1
|
Romero-Sanz S, Caldero-Escudero E, Álvarez-Illera P, Santo-Domingo J, de la Fuente S, García-Casas P, Fonteriz RI, Montero M, Álvarez J. Rescue of a Rotenone Model of Parkinson's Disease in C. elegans by the Mitochondrial Na +/Ca 2+ Exchanger Inhibitor CGP37157. Int J Mol Sci 2025; 26:3371. [PMID: 40244237 PMCID: PMC11989483 DOI: 10.3390/ijms26073371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/24/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
We have previously shown that the compound CGP37157, a mitochondrial Na+/Ca2+ exchanger inhibitor, increases lifespan and improves muscle and mitochondrial structure during aging in wild-type C. elegans nematodes. We used here a rotenone model of Parkinson's disease in C. elegans to test the ability of CGP37157 to rescue the alterations induced by the toxicant. Rotenone, a mitochondrial respiratory chain complex I inhibitor, reduced worm lifespan and muscle activity, measured as worm mobility, pharyngeal pumping, and defecation rate. It also increased ROS production, decreased mitochondrial membrane potential, and disorganized mitochondrial structure. Moreover, it induced degeneration of dopaminergic neurons and changes in behavior. We found that CGP37157 produced a partial or complete reversal of most of these alterations. These results are consistent with our previous proposal that Ca2+ homeostasis is important in the development of neurodegenerative diseases, and modulation of the Ca2+ signaling toolkit may be a novel target for their treatment.
Collapse
Affiliation(s)
- Silvia Romero-Sanz
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain; (S.R.-S.); (E.C.-E.); (P.Á.-I.); (J.S.-D.); (S.d.l.F.); (P.G.-C.); (R.I.F.); (M.M.)
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), 47005 Valladolid, Spain
| | - Elena Caldero-Escudero
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain; (S.R.-S.); (E.C.-E.); (P.Á.-I.); (J.S.-D.); (S.d.l.F.); (P.G.-C.); (R.I.F.); (M.M.)
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), 47005 Valladolid, Spain
| | - Pilar Álvarez-Illera
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain; (S.R.-S.); (E.C.-E.); (P.Á.-I.); (J.S.-D.); (S.d.l.F.); (P.G.-C.); (R.I.F.); (M.M.)
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), 47005 Valladolid, Spain
| | - Jaime Santo-Domingo
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain; (S.R.-S.); (E.C.-E.); (P.Á.-I.); (J.S.-D.); (S.d.l.F.); (P.G.-C.); (R.I.F.); (M.M.)
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), 47005 Valladolid, Spain
| | - Sergio de la Fuente
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain; (S.R.-S.); (E.C.-E.); (P.Á.-I.); (J.S.-D.); (S.d.l.F.); (P.G.-C.); (R.I.F.); (M.M.)
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), 47005 Valladolid, Spain
| | - Paloma García-Casas
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain; (S.R.-S.); (E.C.-E.); (P.Á.-I.); (J.S.-D.); (S.d.l.F.); (P.G.-C.); (R.I.F.); (M.M.)
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), 47005 Valladolid, Spain
| | - Rosalba I. Fonteriz
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain; (S.R.-S.); (E.C.-E.); (P.Á.-I.); (J.S.-D.); (S.d.l.F.); (P.G.-C.); (R.I.F.); (M.M.)
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), 47005 Valladolid, Spain
| | - Mayte Montero
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain; (S.R.-S.); (E.C.-E.); (P.Á.-I.); (J.S.-D.); (S.d.l.F.); (P.G.-C.); (R.I.F.); (M.M.)
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), 47005 Valladolid, Spain
| | - Javier Álvarez
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain; (S.R.-S.); (E.C.-E.); (P.Á.-I.); (J.S.-D.); (S.d.l.F.); (P.G.-C.); (R.I.F.); (M.M.)
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), 47005 Valladolid, Spain
| |
Collapse
|
2
|
Nogueira NDS, Portal TM, Nogueira TDS, Miranda AES, Campos E, Monteiro de Barros C. Neuronal degeneration, mitochondrial dysfunction, and disturbance of movements induced by rotenone in the ascidian Styela plicata. Neurotoxicology 2025; 108:69-80. [PMID: 40074177 DOI: 10.1016/j.neuro.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 03/01/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Parkinson's disease (PD), a movement disorder caused by dopaminergic degeneration in the midbrain, has been induced in various organisms after injection of different neurotoxins, such as rotenone (ROT), which affect mitochondrial complex I. Due to the conserved characteristics of ascidians, these animals constitute an interesting model for comparative and genetic studies of neurodegenerative diseases. In this study, we investigated the effects of ROT on the ascidian nervous system, evaluating apoptosis, catecholaminergic enzymes, behavioral deficits, and mitochondrial dysfunction. The study revealed morphological disorganization, inducing vacuolation in the ascidian brain. Neuronal death was confirmed by elevated transcriptional levels of caspase-3 and intense caspase-3 staining by immunofluorescence. In addition, there was reduced staining for dopa-decarboxylase (DDC), which is involved in dopamine biosynthesis. Furthermore, the mitochondria exhibited dysfunction in their membrane potential, followed by a decrease in the hydrolytic activity of ATP synthase and high transcriptional levels of ubiquitin. Finally, after administration of the drug l-3,4-dihydroxyphenylalanine (L-DOPA), recovery of motor movements was observed, as revealed by behavioral tests. Overall, the current research provides new data on the effects of rotenone on the ascidian brain, inducing neuronal death, mitochondrial dysfunction, and siphon movement disorders in the ascidian Styela plicata.
Collapse
Affiliation(s)
- Nathany da Silva Nogueira
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Macaé 27965-045, Brazil; Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Macaé 27965-045, Brazil.
| | - Taynan Motta Portal
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Macaé 27965-045, Brazil; Programa de Pós-graduação em Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro - UENF, Campo dos Goytacazes, Rio de Janeiro 28013-602, Brazil.
| | - Thuany da Silva Nogueira
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Macaé 27965-045, Brazil; Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Macaé 27965-045, Brazil.
| | - Aurenita Emile Sá Miranda
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Macaé 27965-045, Brazil.
| | - Eldo Campos
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Macaé 27965-045, Brazil; Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Macaé 27965-045, Brazil.
| | - Cintia Monteiro de Barros
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Macaé 27965-045, Brazil; Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Macaé 27965-045, Brazil.
| |
Collapse
|
3
|
Chen X, Liu L, Jiang W, Hu Y, Zou W, Zhang P, Wang B. Hydrogen Sulfide Inhibits Ferritinophagy-Mediated Ferroptosis in the Hippocampus of Rotenone-Exposed Rats. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2025; 68:91-99. [PMID: 40103451 DOI: 10.4103/ejpi.ejpi-d-24-00099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 02/03/2025] [Indexed: 03/20/2025]
Abstract
ABSTRACT Our previous research has established that hydrogen sulfide (H 2 S) exerts an antagonistic effect against the hippocampal neurotoxicity induced by Rotenone (ROT). However, the underlying mechanisms are so far poorly understood. Substantial evidence corroborates the involvement of ferroptosis in ROT-induced neurotoxicity. To elucidate the protective mechanism of H 2 S against ROT-induced hippocampal neurotoxicity, this study explores its regulatory role in ferroptosis and its underlying mechanisms. We used Fluoro-Jade B staining to detect dead neurons. The levels of ferrous ions and glutathione (GSH) were measured by a kit. The ferroptosis-related proteins, including light-chain subunit (xCT), GSH peroxidase 4(GPX4), ferroptosis marker acyl-CoA synthetase long-chain family member 4(ACSL4), and ferritinophagy-related protein, including ferritin heavy chain 1 (FTH1), sequestosome 1 (p62), ferritinophagy markers autophagosome marker light-chain I/II (LC3I/II), and nuclear receptor coactivator 4 (NCOA4), were measured by Western blot. Our findings indicate that H 2 S reduces hippocampal neuron deaths in ROT-exposed rats. Meanwhile, H 2 S reverses the downregulations of xCT and GPX4, and the upregulations of ferrous ion and ACSL4 in the hippocampus induced by ROT. Furthermore, H 2 S reverses the upregulations of LC3I/II and NCOA4, and the downregulations of P62 and FTH1. Based on these findings, we concluded that the protective role of H 2 S against ROT-induced hippocampal neuronal death involves inhibiting ferroptosis triggered by ferritinophagy.
Collapse
Affiliation(s)
- Xi Chen
- Department of Neurology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li Liu
- Department of Neurology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wu Jiang
- Department of Neurology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yu Hu
- Institute of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wei Zou
- Department of Neurology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ping Zhang
- Department of Neurology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Institute of Neuroscience, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Bo Wang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyan, Hunan, China
| |
Collapse
|
4
|
Zhou L, Cai X, Wang Y, Yang J, Wang Y, Deng J, Ye D, Zhang L, Liu Y, Ma S. Chemistry and biology of natural stilbenes: an update. Nat Prod Rep 2025; 42:359-405. [PMID: 39711130 DOI: 10.1039/d4np00033a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Covering: 2009 up to the end of 2023Stilbenes, an emblematic group of polyphenols, have attracted the attention of numerous researchers owing to their intriguing polycyclic architectures and diverse bioactivities. In this updated review, natural stilbenes were analysed, especially oligomeric stilbenes, which are an emblematic group of polyphenols that harbor intriguing polycyclic architectures and diverse bioactivities compared with those previously anticipated. Oligomeric stilbenes with unique skeletons comprise a large majority of natural stilbenes owing to their structural changes and different substitutions on the phenyl rings. These compounds can be promising sources of lead compounds for studying new drugs and medicines. In addition, the exploration of unusual structures of oligomeric stilbenes such as polyflavanostilbenes A and B, analysing their absolute stereochemistry, and improving their yield using synthetic biology methods have recently gained interest. This review provides a systematic overview of 409 new stilbenes, which were isolated and identified over time from January 2009 to December 2023, focusing on the classification and biomimetic syntheses of oligomeric stilbenes, in addition to presenting meaningful insights into their structural diversity and biological activities, which will inspire further investigations of biological activities, structure-activity relationships, and screening of drug candidates.
Collapse
Affiliation(s)
- Lipeng Zhou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xinyu Cai
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Wang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing 100050, China
| | - Jianbo Yang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing 100050, China
| | - Yadan Wang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing 100050, China
| | - Jialing Deng
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Danni Ye
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Lanzhen Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Yue Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Shuangcheng Ma
- Chinese Pharmacopoeia Commission, Beijing 100061, China.
| |
Collapse
|
5
|
Buneeva O, Medvedev A. Monoamine Oxidase Inhibitors in Toxic Models of Parkinsonism. Int J Mol Sci 2025; 26:1248. [PMID: 39941014 PMCID: PMC11818313 DOI: 10.3390/ijms26031248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 02/16/2025] Open
Abstract
Monoamine oxidase inhibitors are widely used for the symptomatic treatment of Parkinson's disease (PD). They demonstrate antiparkinsonian activity in different toxin-based models induced by 6-hydroxydopamine, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and pesticides (rotenone and paraquat). In some models, such as MPTP-induced PD, MAO inhibitors prevent the formation of the neurotoxin MPP+ from the protoxin MPTP. Regardless of the toxin's nature, potent MAO inhibitors prevent dopamine loss reduction, the formation of hydrogen peroxide, hydrogen peroxide signaling, and the accumulation of hydrogen peroxide-derived reactive oxygen species responsible for the development of oxidative stress. It becomes increasingly clear that some metabolites of MAO inhibitors (e.g., the rasagiline metabolite 1-R-aminoindan) possess their own bio-pharmacological activities unrelated to the parent compound. In addition, various MAO inhibitors exhibit multitarget action, in which MAO-independent effects prevail. This opens new prospects in the development of novel therapeutics based on simultaneous actions on several prospective targets for the therapy of PD.
Collapse
Affiliation(s)
| | - Alexei Medvedev
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, 119121 Moscow, Russia;
| |
Collapse
|
6
|
Yashooa RK, Duranti E, Conconi D, Lavitrano M, Mustafa SA, Villa C. Mitochondrial microRNAs: Key Drivers in Unraveling Neurodegenerative Diseases. Int J Mol Sci 2025; 26:626. [PMID: 39859339 PMCID: PMC11766038 DOI: 10.3390/ijms26020626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/07/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs (ncRNAs) crucial for regulating gene expression at the post-transcriptional level. Recent evidence has shown that miRNAs are also found in mitochondria, organelles that produce energy in the cell. These mitochondrial miRNAs, also known as mitomiRs, are essential for regulating mitochondrial function and metabolism. MitomiRs can originate from the nucleus, following traditional miRNA biogenesis pathways, or potentially from mitochondrial DNA, allowing them to directly affect gene expression and cellular energy dynamics within the mitochondrion. While miRNAs have been extensively investigated, the function and involvement of mitomiRs in the development of neurodegenerative disorders like Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis remain to be elucidated. This review aims to discuss findings on the role of mitomiRs in such diseases and their potential as therapeutic targets, as well as to highlight future research directions.
Collapse
Affiliation(s)
- Raya Kh. Yashooa
- Department of Biology, College of Education for Pure Science, University of Al-Hamdaniya, Mosul 41002, Iraq;
| | - Elisa Duranti
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (E.D.); (D.C.); (M.L.)
| | - Donatella Conconi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (E.D.); (D.C.); (M.L.)
| | - Marialuisa Lavitrano
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (E.D.); (D.C.); (M.L.)
| | - Suhad A. Mustafa
- General Directorate of Scientific Research Center, Salahaddin University-Erbil, Kurdistan Region, Erbil 44001, Iraq;
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (E.D.); (D.C.); (M.L.)
| |
Collapse
|
7
|
Silva MAP, Carvalho LIM, Destro MV, Braz LG, Braz MG. From indoors to outdoors: Impact of waste anesthetic gases on occupationally exposed professionals and related environmental hazards - A narrative review and update. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2025; 113:104624. [PMID: 39732353 DOI: 10.1016/j.etap.2024.104624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/19/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Waste anesthetic gases (WAGs) are trace-concentration inhaled anesthetics that exist worldwide because they are released into the ambient air of operating rooms (ORs) and post-anesthesia care units. WAGs cause indoor contamination, especially in ORs lacking proper scavenging systems, and occupational exposure, while promoting climate change through greenhouse gas/ozone-depleting effects. Despite these controversial features, WAGs continue to pose occupational health hazards. Occupational exposure to WAGs has been linked to oxidative stress and cytotoxic, genotoxic and mutagenic potential. This review aims to analyze and update the literature on WAG monitoring, the impact of WAGs on occupationally exposed personnel and their effect on the environment. The awareness of exposed professionals in human and veterinary medicine is crucial. The implementation of biomonitoring practices and WAG occupational exposure limiting policies is needed. Promoting a sustainable healthcare system is also important for mitigating the impact of WAGs on global warming.
Collapse
Affiliation(s)
- Mariane A P Silva
- São Paulo State University (UNESP), Medical School, Division of Anesthesiology, GENOTOX Lab., Botucatu, São Paulo, Brazil
| | - Lorena I M Carvalho
- Getúlio Vargas Hospital, Teresina, Piauí, Brazil; Hospital Unimed Primavera, Department of Anesthesiology, Teresina, Piauí, Brazil
| | - Maria Vitória Destro
- São Paulo State University (UNESP), Medical School, Division of Anesthesiology, GENOTOX Lab., Botucatu, São Paulo, Brazil
| | - Leandro G Braz
- São Paulo State University (UNESP), Medical School, Division of Anesthesiology, GENOTOX Lab., Botucatu, São Paulo, Brazil
| | - Mariana G Braz
- São Paulo State University (UNESP), Medical School, Division of Anesthesiology, GENOTOX Lab., Botucatu, São Paulo, Brazil.
| |
Collapse
|
8
|
Louati K, Maalej A, Kolsi F, Kallel R, Gdoura Y, Borni M, Hakim LS, Zribi R, Choura S, Sayadi S, Chamkha M, Mnif B, Khemakhem Z, Boudawara TS, Boudawara MZ, Bouraoui A, Kraiem J, Safta F. A Shotgun Proteomic-Based Approach with a Q-Exactive Hybrid Quadrupole-Orbitrap High-Resolution Mass Spectrometer for the Assessment of Pesticide Mixture-Induced Neurotoxicity on a 3D-Developed Neurospheroid Model from Human Brain Meningiomas: Identification of Trityl-Post-Translational Modification. J Proteome Res 2024; 23:5554-5576. [PMID: 39556108 PMCID: PMC11629387 DOI: 10.1021/acs.jproteome.4c00804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/14/2024] [Accepted: 10/31/2024] [Indexed: 11/19/2024]
Abstract
The widespread use of pesticides, particularly in combinations, has resulted in enhanced hazardous health effects. However, little is known about their molecular mechanism of interactions. The aim of this study was to assess the neurotoxicity effect of pesticides in mixtures by adopting a 3D in vitro developed neurospheroid model, followed by treatment by increased concentrations of pesticides for 24 h and analysis by a shotgun proteomic-based approach with high-resolution tandem mass spectrometry. Three proteins, namely, glyceraldehyde-3-phosphate-dehydrogenase (GAPDH), α-enolase, and phosphoglycerate-kinase-1, were selected as key targets in the metabolic process. Only high doses of pesticides mitigated cell-density proliferation with the occurrence of apoptotic cells, which unlikely makes any neurological alterations in environmental regulatory exposures. The proteomic analysis showed that majority of altered proteins were implicated in cell metabolism. De novo peptide sequencing revealed ion losses and adduct formation, namely, a trityl-post-translational modification in the active site of 201-GAPDH protein. The study also highlights the plausible role of pyrethroids to be implicated in the deleterious effects of pesticides in a mixture. To the best of our knowledge, our finding is the first in toxicoproteomics to deeply elucidate pesticides' molecular interactions and their ability to adduct proteins as a pivotal role in the neurotoxicity mechanism.
Collapse
Affiliation(s)
- Kaouthar Louati
- Laboratory
of Chemical, Galenic and Pharmacological Drug Development- LR12ES09, University of Monastir, Road Avicenne , 5000Monastir, Tunisia
| | - Amina Maalej
- Laboratory
of Environmental Bioprocesses, Centre of
Biotechnology of Sfax, Road of Sidi-Mansour, P.O. Box 1177 , 3018Sfax, Tunisia
| | - Fatma Kolsi
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Rim Kallel
- Laboratory
of Pathological Anatomy and Cytology, Habib
Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Yassine Gdoura
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Mahdi Borni
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Leila Sellami Hakim
- Laboratory
of Pathological Anatomy and Cytology, Habib
Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
| | - Rania Zribi
- Faculty
of Letters and Humanities, University of
Sfax, Airport Road, Km
4.5, 3023 Sfax, Tunisia
| | - Sirine Choura
- Laboratory
of Environmental Bioprocesses, Centre of
Biotechnology of Sfax, Road of Sidi-Mansour, P.O. Box 1177 , 3018Sfax, Tunisia
| | - Sami Sayadi
- Biotechnology
Program, Center for Sustainable Development, College of Arts and Sciences, Qatar University, 2713 Doha, Qatar
| | - Mohamed Chamkha
- Laboratory
of Environmental Bioprocesses, Centre of
Biotechnology of Sfax, Road of Sidi-Mansour, P.O. Box 1177 , 3018Sfax, Tunisia
| | - Basma Mnif
- Department
of Bacteriology, Habib Bourguiba University
Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Zouheir Khemakhem
- Legal
Medicine
Department, Habib Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Tahya Sellami Boudawara
- Laboratory
of Pathological Anatomy and Cytology, Habib
Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Mohamed Zaher Boudawara
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Abderrahman Bouraoui
- Laboratory
of Chemical, Galenic and Pharmacological Drug Development- LR12ES09, University of Monastir, Road Avicenne , 5000Monastir, Tunisia
| | - Jamil Kraiem
- Laboratory
of Chemical, Galenic and Pharmacological Drug Development- LR12ES09, University of Monastir, Road Avicenne , 5000Monastir, Tunisia
| | - Fathi Safta
- Laboratory
of Chemical, Galenic and Pharmacological Drug Development- LR12ES09, University of Monastir, Road Avicenne , 5000Monastir, Tunisia
| |
Collapse
|
9
|
Zhang F, Ye Z, Xie Y, Liu M, Zhang L, Zhang J, Xu Z. Levodopa-induced dyskinesia: brain iron deposition as a new hypothesis. Biometals 2024; 37:1307-1323. [PMID: 39212870 DOI: 10.1007/s10534-024-00628-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease in the older adults. The main pathological change in PD is the degenerative death of dopamine (DA) neurons in the midbrain substantia nigra, which causes a significant decrease in the DA content of the striatum. However, the exact etiology of this pathological change remains unclear. Genetic factors, environmental factors, aging, and oxidative stress may be involved in the degenerative death of dopaminergic neurons in PD. Pharmacological treatment using levodopa (L-DOPA) remains the main treatment for PD. Most patients with PD consuming L-DOPA for a long time usually develop levodopa-induced dyskinesia (LID) after 6.5 years of use, and LID seriously affects the quality of life and increases the risk of disability. Recently, studies have revealed that cerebral iron deposition may be involved in LID development and that iron deposition has neurotoxic effects and accelerates disease onset. However, the relationship between cerebral iron deposition and LID remains unclear. Herein, we reviewed the mechanisms by which iron deposition may be associated with LID development, which are mainly related to oxidative stress, neuroinflammation, and mitochondrial and lysosomal dysfunction. Using iron as an important target, the search and development of safe and effective brain iron scavengers, and thus the alleviation and treatment of LID, has a very important scientific and clinical value, as well as a good application prospect.
Collapse
Affiliation(s)
- Fanshi Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Zhuofan Ye
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
- Department of Neurology, Guizhou Pronvincial People's Hospital, Guiyang, China
| | - Yuanyang Xie
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Mei Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Li Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Jun Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China.
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
10
|
Wu Z, Zhao T, Jiang X, Zhang D, Wang F, Ren X, Wang Z, Wang E, Ren J. A near-infrared fluorescent probe with a large Stokes shift for the detection and imaging of biothiols in vitro and in vivo. Anal Bioanal Chem 2024; 416:6485-6495. [PMID: 39322801 DOI: 10.1007/s00216-024-05537-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/18/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024]
Abstract
In this study, a new near-infrared (NIR) fluorescent turn-on probe featuring a large Stokes shift (198 nm) was developed for the detection of biothiols. The probe was based on a dicyanoisophorone derivative serving as the fluorophore and a 2,4-dinitrobenzenesulfonyl (DNBS) group functioning as both a recognition site and a fluorescence quencher. In the absence of biothiols, the fluorescence of the probe was low due to the photoinduced electron transfer (PET) effect between the fluorophore and DNBS. Upon the presence of biothiols, the DNBS group underwent a nucleophilic aromatic substitution reaction with the sulfhydryl group of biothiols, leading to the release of the fluorophore and a notable emission peak at 668 nm. This developed probe exhibited exceptional selectivity and sensitivity to biothiols in solution, with an impressive detection limit of 28 nM for cysteine (Cys), 22 nM for homocysteine (Hcy), and 24 nM for glutathione (GSH). Furthermore, the probe demonstrated its applicability by successfully visualizing both endogenous and exogenous biothiols in living systems.
Collapse
Affiliation(s)
- Zhengjun Wu
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan, 430062, People's Republic of China
| | - Taotao Zhao
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan, 430062, People's Republic of China
| | - Xingyue Jiang
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan, 430062, People's Republic of China
| | - Dan Zhang
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan, 430062, People's Republic of China
| | - Feiyi Wang
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan, 430062, People's Republic of China
| | - Xiaoming Ren
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan, 430062, People's Republic of China
| | - Zhao Wang
- Wuhan Business University, Wuhan, 430056, People's Republic of China.
| | - Erfei Wang
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan, 430062, People's Republic of China.
| | - Jun Ren
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan, 430062, People's Republic of China.
| |
Collapse
|
11
|
Zarei P, Amirpour-Najafabadi B, Sam-Sani P, Sakhaie MH, Sadegh M. Protective Effect of Whey Protein Supplement Against Rotenone Induced Motor Dysfunction in a Rat Model of Parkinson Disease. Adv Biomed Res 2024; 13:93. [PMID: 39717252 PMCID: PMC11665149 DOI: 10.4103/abr.abr_178_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/16/2023] [Accepted: 09/23/2023] [Indexed: 12/25/2024] Open
Abstract
Background We aimed to investigate the effects of whey protein (WP) supplements in a rat model of rotenone-induced locomotor and biochemical features of Parkinson's disease (PD). Materials and Methods Male Wistar rats were used. Daily injections of rotenone (2 mg/kg; i.p.) for 16 days were used to induce PD. WP or soy protein (SP) at 1, 2, and 4 g/rats were administrated daily by gavage. Motor skills were measured in rats 24 h after the last injection using the bar test, grid test, rearing, and open field tests. In the following, striatum tissue was isolated for biochemical measurements. ELISA kits were used for biochemical assessments. Results While rotenone caused a significant increase in the delay time in both the bar and grid tests and a significant decrease in the motor activities were observed in both rearing and spontaneous movement tests in the rotenone group, supplementation with 2 and 4 g of WP, but not SP, significantly decreased the delay time in the bar and grid tests and also significantly increased both rearing and spontaneous movements. Additionally, rotenone caused a significant decrease in striatal levels of dopamine and glutathione and significantly increased apoptotic caspases 8, 9, and Cytochrome C, while 2 and 4 g of WP, but not SP, significantly reversed these effects. Conclusions WP appears to have neuroprotective effects against rotenone-induced neurotoxicity and motor dysfunction, so it may be effective in the control of PD.
Collapse
Affiliation(s)
- Parvin Zarei
- Department of Bioinformatics, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Parnian Sam-Sani
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | | | - Mehdi Sadegh
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
12
|
Neha, Mazahir I, Khan SA, Kaushik P, Parvez S. The Interplay of Mitochondrial Bioenergetics and Dopamine Agonists as an Effective Disease-Modifying Therapy for Parkinson's Disease. Mol Neurobiol 2024; 61:8086-8103. [PMID: 38468113 DOI: 10.1007/s12035-024-04078-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/28/2024] [Indexed: 03/13/2024]
Abstract
Parkinson's disease (PD) is a progressive neurological ailment with a slower rate of advancement that is more common in older adults. The biggest risk factor for PD is getting older, and those over 60 have an exponentially higher incidence of this condition. The failure of the mitochondrial electron chain, changes in the dynamics of the mitochondria, and abnormalities in calcium and ion homeostasis are all symptoms of Parkinson's disease (PD). Increased mitochondrial reactive oxygen species (mROS) and an energy deficit are linked to these alterations. Levodopa (L-DOPA) is a medication that is typically used to treat most PD patients, but because of its negative effects, additional medications have been created utilizing L-DOPA as the parent molecule. Ergot and non-ergot derivatives make up most PD medications. PD is successfully managed with the use of dopamine agonists (DA). To get around the motor issues produced by L-DOPA, these dopamine derivatives can directly excite DA receptors in the postsynaptic membrane. In the past 10 years, two non-ergoline DA with strong binding properties for the dopamine D2 receptor (D2R) and a preference for the dopamine D3 receptor (D3R) subtype, ropinirole, and pramipexole (PPx) have been developed for the treatment of PD. This review covers the most recent research on the efficacy and safety of non-ergot drugs like ropinirole and PPx as supplementary therapy to DOPA for the treatment of PD.
Collapse
Affiliation(s)
- Neha
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Iqra Mazahir
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Sara Akhtar Khan
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Pooja Kaushik
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
13
|
Avallone R, Rustichelli C, Filaferro M, Vitale G. Chemical Characterization and Beneficial Effects of Walnut Oil on a Drosophila melanogaster Model of Parkinson's Disease. Molecules 2024; 29:4190. [PMID: 39275038 PMCID: PMC11397333 DOI: 10.3390/molecules29174190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 09/16/2024] Open
Abstract
A nutritional approach could be a promising strategy to prevent or decrease the progression of neurodegenerative disorders such as Parkinson's disease (PD). The neuroprotective role of walnut oil (WO) was investigated in Drosophila melanogaster treated with rotenone (Rot), as a PD model, WO, or their combination, and compared to controls. WO reduced mortality and improved locomotor activity impairment after 3 and 7 days, induced by Rot. LC-MS analyses of fatty acid levels in Drosophila heads showed a significant increase in linolenic (ALA) and linoleic acid (LA) both in flies fed with the WO-enriched diet and in those treated with the association of WO with Rot. Flies supplemented with the WO diet showed an increase in brain dopamine (DA) level, while Rot treatment significantly depleted dopamine content; conversely, the association of Rot with WO did not modify DA content compared to controls. The greater intake of ALA and LA in the enriched diet enhanced their levels in Drosophila brain, suggesting a neuroprotective role of polyunsaturated fatty acids against Rot-induced neurotoxicity. The involvement of the dopaminergic system in the improvement of behavioral and biochemical parameters in Drosophila fed with WO is also suggested.
Collapse
Affiliation(s)
- Rossella Avallone
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Cecilia Rustichelli
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Monica Filaferro
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giovanni Vitale
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
14
|
Pullara F, Forsmann MC, General IJ, Ayoob JC, Furbee E, Castro SL, Hu X, Greenamyre JT, Di Maio R. NADPH oxidase 2 activity disrupts Calmodulin/CaMKIIα complex via redox modifications of CaMKIIα-contained Cys30 and Cys289: Implications in Parkinson's disease. Redox Biol 2024; 75:103254. [PMID: 38968922 PMCID: PMC11278932 DOI: 10.1016/j.redox.2024.103254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/12/2024] [Accepted: 06/22/2024] [Indexed: 07/07/2024] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II α (CaMKIIα) signaling in the brain plays a critical role in regulating neuronal Ca2+ homeostasis. Its dysfunctional activity is associated with various neurological and neurodegenerative disorders, including Parkinson's disease (PD). Using computational modeling analysis, we predicted that, two essential cysteine residues contained in CaMKIIα, Cys30 and Cys289, may undergo redox modifications impacting the proper functioning of the CaMKIIα docking site for Ca2+/CaM, thus impeding the formation of the CaMKIIα:Ca2+/CaM complex, essential for a proper modulation of CaMKIIα kinase activity. Our subsequent in vitro investigations confirmed the computational predictions, specifically implicating Cys30 and Cys289 residues in impairing CaMKIIα:Ca2+/CaM interaction. We observed CaMKIIα:Ca2+/CaM complex disruption in dopamine (DA) nigrostriatal neurons of post-mortem Parkinson's disease (PD) patients' specimens, addressing the high relevance of this event in the disease. CaMKIIα:Ca2+/CaM complex disruption was also observed in both in vitro and in vivo rotenone models of PD, where this phenomenon was associated with CaMKIIα kinase hyperactivity. Moreover, we observed that, NADPH oxidase 2 (NOX2), a major enzymatic generator of superoxide anion (O2●-) and hydrogen peroxide (H2O2) in the brain with implications in PD pathogenesis, is responsible for CaMKIIα:Ca2+/CaM complex disruption associated to a stable Ca2+CAM-independent CaMKIIα kinase activity and intracellular Ca2+ accumulation. The present study highlights the importance of oxidative stress, in disturbing the delicate balance of CaMKIIα signaling in calcium dysregulation, offering novel insights into PD pathogenesis.
Collapse
Affiliation(s)
| | - Madison C Forsmann
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA
| | - Ignacio J General
- School of Science and Technology, Universidad Nacional de San Martin, San Martín, 1650, Buenos Aires, Argentina
| | - Joseph C Ayoob
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Emily Furbee
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Sandra L Castro
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA
| | - Xiaoping Hu
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA
| | - J Timothy Greenamyre
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA
| | - Roberto Di Maio
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
15
|
Badae NM, Abdelmonsif DA, Aly RG, Omar AM, Shoela MS, Omar EM. Effect of spermidine on long non-coding RNAs MALAT1 in a rotenone induced-rat model of Parkinson's disease. Fundam Clin Pharmacol 2024; 38:718-729. [PMID: 38279557 DOI: 10.1111/fcp.12986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 12/19/2023] [Accepted: 01/08/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Spermidine is a natural biologically active substance that has widespread influences on the body. OBJECTIVE This study aims to enhance our understanding of the potential effect of spermidine on long non-coding RNA MALAT1 and explore the underlying mechanism in the rotenone-induced rat model of Parkinson's disease. METHODS Rats were sacrificed after locomotor behavioral testing. Striatal tissues were used to assess the expression of MALAT1, oxidative stress markers, and autophagy markers. RESULTS Our study found that treatment with spermidine for 2 weeks during the induction of the model significantly improved behavioral assessment, dopamine levels, and attenuated the histopathological changes that occurred in PD in comparison to the non-treated group. CONCLUSION Our preliminary study supports the protective effect of spermidine on the activation of autophagy and its antioxidant properties. Part of the antioxidant activity is due to the inhibition of MALAT1. However, MALAT1 does not correlate with the spermidine-induced autophagy pathway.
Collapse
Affiliation(s)
- Noha Mohamed Badae
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Doaa A Abdelmonsif
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Rania Gaber Aly
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Amira M Omar
- Department of Histology & Cell Biology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mai S Shoela
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Eman M Omar
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
16
|
Bucher ML, Dicent J, Duarte Hospital C, Miller GW. Neurotoxicology of dopamine: Victim or assailant? Neurotoxicology 2024; 103:175-188. [PMID: 38857676 PMCID: PMC11694735 DOI: 10.1016/j.neuro.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
Since the identification of dopamine as a neurotransmitter in the mid-20th century, investigators have examined the regulation of dopamine homeostasis at a basic biological level and in human disorders. Genetic animal models that manipulate the expression of proteins involved in dopamine homeostasis have provided key insight into the consequences of dysregulated dopamine. As a result, we have come to understand the potential of dopamine to act as an endogenous neurotoxin through the generation of reactive oxygen species and reactive metabolites that can damage cellular macromolecules. Endogenous factors, such as genetic variation and subcellular processes, and exogenous factors, such as environmental exposures, have been identified as contributors to the dysregulation of dopamine homeostasis. Given the variety of dysregulating factors that impact dopamine homeostasis and the potential for dopamine itself to contribute to further cellular dysfunction, dopamine can be viewed as both the victim and an assailant of neurotoxicity. Parkinson's disease has emerged as the exemplar case study of dopamine dysregulation due to the genetic and environmental factors known to contribute to disease risk, and due to the evidence of dysregulated dopamine as a pathologic and pathogenic feature of the disease. This review, inspired by the talk, "Dopamine in Durham: location, location, location" presented by Dr. Miller for the Jacob Hooisma Memorial Lecture at the International Neurotoxicology Association meeting in 2023, offers a primer on dopamine toxicity covering endogenous and exogenous factors that disrupt dopamine homeostasis and the actions of dopamine as an endogenous neurotoxin.
Collapse
Affiliation(s)
- Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Jocelyn Dicent
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Carolina Duarte Hospital
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA; Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
17
|
Currim F, Tanwar R, Brown-Leung JM, Paranjape N, Liu J, Sanders LH, Doorn JA, Cannon JR. Selective dopaminergic neurotoxicity modulated by inherent cell-type specific neurobiology. Neurotoxicology 2024; 103:266-287. [PMID: 38964509 PMCID: PMC11288778 DOI: 10.1016/j.neuro.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative disease affecting millions of individuals worldwide. Hallmark features of PD pathology are the formation of Lewy bodies in neuromelanin-containing dopaminergic (DAergic) neurons of the substantia nigra pars compacta (SNpc), and the subsequent irreversible death of these neurons. Although genetic risk factors have been identified, around 90 % of PD cases are sporadic and likely caused by environmental exposures and gene-environment interaction. Mechanistic studies have identified a variety of chemical PD risk factors. PD neuropathology occurs throughout the brain and peripheral nervous system, but it is the loss of DAergic neurons in the SNpc that produce many of the cardinal motor symptoms. Toxicology studies have found specifically the DAergic neuron population of the SNpc exhibit heightened sensitivity to highly variable chemical insults (both in terms of chemical structure and mechanism of neurotoxic action). Thus, it has become clear that the inherent neurobiology of nigral DAergic neurons likely underlies much of this neurotoxic response to broad insults. This review focuses on inherent neurobiology of nigral DAergic neurons and how such neurobiology impacts the primary mechanism of neurotoxicity. While interactions with a variety of other cell types are important in disease pathogenesis, understanding how inherent DAergic biology contributes to selective sensitivity and primary mechanisms of neurotoxicity is critical to advancing the field. Specifically, key biological features of DAergic neurons that increase neurotoxicant susceptibility.
Collapse
Affiliation(s)
- Fatema Currim
- School of Health Sciences, Purdue University, West Lafayette, IN 47901, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47901, USA
| | - Reeya Tanwar
- School of Health Sciences, Purdue University, West Lafayette, IN 47901, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47901, USA
| | - Josephine M Brown-Leung
- School of Health Sciences, Purdue University, West Lafayette, IN 47901, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47901, USA
| | - Neha Paranjape
- Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Jennifer Liu
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC 27710, USA; Duke Center for Neurodegeneration and Neurotherapeutics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Laurie H Sanders
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC 27710, USA; Duke Center for Neurodegeneration and Neurotherapeutics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jonathan A Doorn
- Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Jason R Cannon
- School of Health Sciences, Purdue University, West Lafayette, IN 47901, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47901, USA.
| |
Collapse
|
18
|
Khafajah Y, Shaheen M, Natour DE, Merheb M, Matar R, Borjac J. Neuroprotective Effects of Zinc Oxide Nanoparticles in a Rotenone-Induced Mouse Model of Parkinson's Disease. Nanotheranostics 2024; 8:497-505. [PMID: 38961888 PMCID: PMC11217785 DOI: 10.7150/ntno.95863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/11/2024] [Indexed: 07/05/2024] Open
Abstract
Goals of the investigation: This work aimed to evaluate the neuroprotective effects of zinc oxide (ZnO) nanoparticles in an experimental mouse model of rotenone-induced PD and investigate the therapeutic effects of ZnO, cobalt ferrite nanoparticles, and their combination. Methods: The levels of dopamine, norepinephrine, epinephrine, and serotonin were assessed using ELISA in the control and experimental model of PD mice. The dopa-decarboxylase expression level was assayed by real-time PCR. The expression level of tyrosine hydroxylase (TH) was assessed by western blot analysis. Results: Our data showed that levels of dopamine decreased in PD mice compared to normal. ZnO NP increased dopamine levels in normal and PD mice (37.5% and 29.5%; respectively, compared to untreated mice). However, ZnO NP did not cause any change in norepinephrine and epinephrine levels either in normal or in PD mice. Levels of serotonin decreased by 64.0%, and 51.1% in PD mice treated with cobalt ferrite and dual ZnO- cobalt ferrite NPs; respectively, when compared to PD untreated mice. The mRNA levels of dopa-decarboxylase increased in both normal and PD mice treated with ZnO NP. Its level decreased when using cobalt ferrite NP and the dual ZnO-cobalt ferrite NP when compared to untreated PD mice. A significant decrease in TH expression by 0.25, 0.68, and 0.62 folds was observed in normal mice treated with ZnO, cobalt ferrite, and the dual ZnO-cobalt ferrite NP as compared to normal untreated mice. In PD mice, ZnO administration caused a non-significant 0.15-fold decrease in TH levels while both cobalt ferrite and the dual ZnO-cobalt ferrite NP administration caused a significant 0.3 and 0.4-fold decrease respectively when compared to untreated PD mice. Principal conclusion: This study reveals that ZnO NPs may be utilized as a potential intervention to elevate dopamine levels to aid in PD treatment.
Collapse
Affiliation(s)
- Yasmeen Khafajah
- Beirut Arab University, Department of Biological Sciences, Faculty of Science, Debbieh, Lebanon
| | - Mariam Shaheen
- Beirut Arab University, Department of Biological Sciences, Faculty of Science, Debbieh, Lebanon
| | - Dania El Natour
- Beirut Arab University, Department of Internal Medicine, Faculty of Medicine, Beirut, Lebanon
| | - Maxime Merheb
- Liwa College, College of Medical and Health Sciences, United Arab Emirates
| | - Rachel Matar
- American University of Ras Al Khaimah, School of Arts and Sciences, United Arab Emirates
| | - Jamilah Borjac
- Beirut Arab University, Department of Biological Sciences, Faculty of Science, Debbieh, Lebanon
| |
Collapse
|
19
|
Mir IH, Anilkumar AS, Guha S, Mohanty AK, Suresh Kumar M, Sujatha V, Ramesh T, Thirunavukkarasu C. Elucidation of 7,8-dihydroxy flavone in complexing with the oxidative stress-inducing enzymes, its impact on radical quenching and DNA damage: an in silico and in vitro approach. J Biomol Struct Dyn 2024; 42:4048-4063. [PMID: 37261742 DOI: 10.1080/07391102.2023.2218932] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/21/2023] [Indexed: 06/02/2023]
Abstract
Oxidative stress (OS) has been attributed to the progression of various disorders, including cancer, diabetes, and cardiovascular diseases. Several antioxidant compounds and free radical quenchers have been shown to mitigate oxidative stress. However, large-scale randomized controlled trials of such compounds on chronic disease aversion have yielded paradoxical and disappointing results due to the constrained cognizance of their oxidative mechanisms and therapeutic targets. The current study sought to identify the potential therapeutic targets of 7,8-Dihydroxyflavone (7,8-DHF) by analyzing its interactions with the enzymes implicated in oxidative stress and also to explore its radicle quenching potential and prophylactic impact on the H2O2-induced DNA damage. Through the in silco approach, we investigated the antioxidant potential of 7,8-DHF by evaluating its interactions with the human oxidative stress-inducing enzymes such as myeloperoxidase (MPO), NADPH oxidase (NOX), nitric oxide synthase (NOS), and xanthine oxidase (XO) and a comparative analysis of those interactions with known antioxidants (Ascorbic acid, Melatonin, Tocopherol) used as controls. The best-scoring complex was adopted for the simulation analysis in investigating protein-ligand conformational dynamics. The in vitro radicle quenching potential was evaluated by performing a spectrum of antioxidant assays, and radical quenching was observed in a dose-dependent fashion with IC50 values of < 60 µM/mL. Further, we probed its anti-hemolytic potential and prophylactic impact in avian erythrocytes subjected to H2O2-induced hemolysis and DNA damage by implementing hemolysis and comet assays. The protective effect was more pronounced at higher concentrations of the drug.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ishfaq Hassan Mir
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | | | - Shreyoshi Guha
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | | | | | - Venugopal Sujatha
- DST-Mobility Fellow, Department of Chemistry, Pondicherry University, Puducherry, India
- Department of Chemistry, Periyar University, Salem, Tamil Nadu, India
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | |
Collapse
|
20
|
Pingale TD, Gupta GL. Oleanolic acid-based therapeutics ameliorate rotenone-induced motor and depressive behaviors in parkinsonian male mice via controlling neuroinflammation and activating Nrf2-BDNF-dopaminergic signaling pathways. Toxicol Mech Methods 2024; 34:335-349. [PMID: 38084769 DOI: 10.1080/15376516.2023.2288198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/21/2023] [Indexed: 04/20/2024]
Abstract
Parkinson's disease (PD) is often accompanied by depression, which may appear before motor signs. Oleanolic acid (OA), a pentacyclic triterpenoid substance, have many pharmacological properties. However, its efficacy in treating PD-related chronic unpredictable stress (CUS) is unknown. Our study used behavioral, biochemical, and immunohistochemical techniques to assess how OA affected PDrelated CUS. Rotenone (1 mg/kg i.p. for first 21 days) was used to induce Parkinsonism, and modest psychological & environmental stresses generated CUS (from day 22 to day 43) in animals. The study included daily i.p.administration of OA (5, 10, and 20 mg/kg) from day 1 to day 57 in male swiss albino mice. Animals were evaluated for behavioral, biochemical parameters, neurotransmitters, and immunohistochemical expression following the treatment. Results of the study revealed that treatment with OA at all doses alleviated the core symptoms of CUS linked to PD and improved motor and non-motor function. OA therapy significantly lowered IL-1β, TNF-α (p < 0.01, < 0.01, < 0.001), IL-6 (p < 0.05, < 0.01, < 0.001), oxidative stress (p < 0.05, < 0.01, < 0.01), and elevated norepinephrine (p < 0.05, < 0.01, < 0.01), dopamine, and serotonin (p < 0.05, < 0.01, < 0.001) levels. Moreover, OA therapy substantially reduced α-synuclein (p < 0.05, < 0.01, < 0.01) aggregation and increased BDNF (p < 0.05, < 0.01, < 0.001) & Nrf-2 (p < 0.05, < 0.01, < 0.01) levels, which boosts neuronal dopamine survival. The study's findings indicated that OA ameliorates depressive-like behavior persuaded by CUS in PD, decreases neuroinflammation, and improves neurotransmitter concentration via activating Nrf2-BDNF-dopaminergic pathway.
Collapse
Affiliation(s)
- Tanvi Dayanand Pingale
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, Vile Parle (W), Mumbai India
| | - Girdhari Lal Gupta
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, Vile Parle (W), Mumbai India
- Department of Pharmacology, School of Pharmacy & Technology Management, SVKM'S Narsee Monjee Institute of Management Studies, Shirpur India
| |
Collapse
|
21
|
Zhang H, Yang J, Guo Y, Lü P, Gong X, Chen K, Li X, Tang M. Rotenone-induced PINK1/Parkin-mediated mitophagy: establishing a silkworm model for Parkinson's disease potential. Front Mol Neurosci 2024; 17:1359294. [PMID: 38706874 PMCID: PMC11066238 DOI: 10.3389/fnmol.2024.1359294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/22/2024] [Indexed: 05/07/2024] Open
Abstract
Parkinson's disease (PD), ranking as the second most prevalent neurodegenerative disorder globally, presents a pressing need for innovative animal models to deepen our understanding of its pathophysiology and explore potential therapeutic interventions. The development of such animal models plays a pivotal role in unraveling the complexities of PD and investigating promising treatment avenues. In this study, we employed transcriptome sequencing on BmN cells treated with 1 μg/ml rotenone, aiming to elucidate the underlying toxicological mechanisms. The investigation brought to light a significant reduction in mitochondrial membrane potential induced by rotenone, subsequently triggering mitophagy. Notably, the PTEN induced putative kinase 1 (PINK1)/Parkin pathway emerged as a key player in the cascade leading to rotenone-induced mitophagy. Furthermore, our exploration extended to silkworms exposed to 50 μg/ml rotenone, revealing distinctive motor dysfunction as well as inhibition of Tyrosine hydroxylase (TH) gene expression. These observed effects not only contribute valuable insights into the impact and intricate mechanisms of rotenone exposure on mitophagy but also provide robust scientific evidence supporting the utilization of rotenone in establishing a PD model in the silkworm. This comprehensive investigation not only enriches our understanding of the toxicological pathways triggered by rotenone but also highlights the potential of silkworms as a valuable model organism for PD research.
Collapse
Affiliation(s)
- Hantao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jinyue Yang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Yinglu Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Peng Lü
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xun Gong
- Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Keping Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiubin Li
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
22
|
Zhang S, Hou J, Zhang X, Cai T, Chen W, Zhang Q. Potential mechanism of biochar enhanced degradation of oxytetracycline by Pseudomonas aeruginosa OTC-T. CHEMOSPHERE 2024; 351:141288. [PMID: 38272135 DOI: 10.1016/j.chemosphere.2024.141288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/11/2023] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Extensive use of oxytetracycline (OTC) and the generation of its corresponding resistance genes have resulted in serious environmental problems. Physical-biological combined remediation is an attractive method for OTC degradation because of its high remediation efficiency, stability, and environmental friendliness. In this study, an effective OTC-degrading strain identified as Pseudomonas aeruginosa OTC-T, was isolated from chicken manure. In the degradation experiment, the degradation rates of OTC in the degradation systems with and without the biochar addition were 92.71-100 % and 69.11-99.59 %, respectively. Biochar improved the tolerance of the strain to extreme environments, and the OTC degradation rate increased by 20.25 %, 18.61 %, and 13.13 % under extreme pH, temperature, and substrate concentration conditions, respectively. Additionally, the degradation kinetics showed that biochar increased the reaction rate constant in the degradation system and shortened the degradation period. In the biological toxicity assessment, biochar increased the proportion of live cells by 17.63 % and decreased the proportion of apoptotic cells by 58.87 %. Metabolomics revealed that biochar had a significant effect on the metabolism of the strains and promoted cell growth and reproduction, effectively reducing oxidative stress induced by OTC. This study elucidates how biochar affects OTC biodegradation and provides insights into the future application of biochar-assisted microbial technology in environmental remediation.
Collapse
Affiliation(s)
- Shudong Zhang
- Shanghai Key Lab for Urban Ecological Processes and Eco-Restoration, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China
| | - Jinju Hou
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Xiaotong Zhang
- Shanghai Key Lab for Urban Ecological Processes and Eco-Restoration, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China
| | - Tong Cai
- Shanghai Key Lab for Urban Ecological Processes and Eco-Restoration, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China
| | - Wenjie Chen
- Shanghai Key Lab for Urban Ecological Processes and Eco-Restoration, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China
| | - Qiuzhuo Zhang
- Shanghai Key Lab for Urban Ecological Processes and Eco-Restoration, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China; Institute of Eco-Chongming (IEC), 3663 N. Zhongshan Rd., Shanghai 200062, China; Technology Innovation Center for Land Spatial Eco-restoration in Metropolitan Area, Ministry of Natural Resources, 3663 N. Zhongshan Road, Shanghai 200062, China.
| |
Collapse
|
23
|
Ay M, Charli A, Langley M, Jang A, Padhi P, Jin H, Anantharam V, Kalyanaraman B, Kanthasamy A, Kanthasamy AG. Mito-metformin protects against mitochondrial dysfunction and dopaminergic neuronal degeneration by activating upstream PKD1 signaling in cell culture and MitoPark animal models of Parkinson's disease. Front Neurosci 2024; 18:1356703. [PMID: 38449738 PMCID: PMC10915001 DOI: 10.3389/fnins.2024.1356703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/07/2024] [Indexed: 03/08/2024] Open
Abstract
Impaired mitochondrial function and biogenesis have strongly been implicated in the pathogenesis of Parkinson's disease (PD). Thus, identifying the key signaling mechanisms regulating mitochondrial biogenesis is crucial to developing new treatment strategies for PD. We previously reported that protein kinase D1 (PKD1) activation protects against neuronal cell death in PD models by regulating mitochondrial biogenesis. To further harness the translational drug discovery potential of targeting PKD1-mediated neuroprotective signaling, we synthesized mito-metformin (Mito-Met), a mitochondria-targeted analog derived from conjugating the anti-diabetic drug metformin with a triphenylphosphonium functional group, and then evaluated the preclinical efficacy of Mito-Met in cell culture and MitoPark animal models of PD. Mito-Met (100-300 nM) significantly activated PKD1 phosphorylation, as well as downstream Akt and AMPKα phosphorylation, more potently than metformin, in N27 dopaminergic neuronal cells. Furthermore, treatment with Mito-Met upregulated the mRNA and protein expression of mitochondrial transcription factor A (TFAM) implying that Mito-Met can promote mitochondrial biogenesis. Interestingly, Mito-Met significantly increased mitochondrial bioenergetics capacity in N27 dopaminergic cells. Mito-Met also reduced mitochondrial fragmentation induced by the Parkinsonian neurotoxicant MPP+ in N27 cells and protected against MPP+-induced TH-positive neurite loss in primary neurons. More importantly, Mito-Met treatment (10 mg/kg, oral gavage for 8 week) significantly improved motor deficits and reduced striatal dopamine depletion in MitoPark mice. Taken together, our results demonstrate that Mito-Met possesses profound neuroprotective effects in both in vitro and in vivo models of PD, suggesting that pharmacological activation of PKD1 signaling could be a novel neuroprotective translational strategy in PD and other related neurocognitive diseases.
Collapse
Affiliation(s)
- Muhammet Ay
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
| | - Adhithiya Charli
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
| | - Monica Langley
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
| | - Ahyoung Jang
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | - Piyush Padhi
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | - Huajun Jin
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | - Vellareddy Anantharam
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | | | - Arthi Kanthasamy
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | - Anumantha G. Kanthasamy
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| |
Collapse
|
24
|
Mohammed HS, Hosny EN, Sawie HG, Khadrawy YA. Transcranial photobiomodulation ameliorates midbrain and striatum neurochemical impairments and behavioral deficits in reserpine-induced parkinsonism in rats. Photochem Photobiol Sci 2023; 22:2891-2904. [PMID: 37917308 DOI: 10.1007/s43630-023-00497-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/15/2023] [Indexed: 11/04/2023]
Abstract
Photobiomodulation (PBM) of deep brain structures through transcranial infrared irradiation might be an effective treatment for Parkinson's disease (PD). However, the mechanisms underlying this intervention should be elucidated to optimize the therapeutic outcome and maximize therapeutic efficacy. The present study aimed at investigating the oxidative stress-related parameters of malondialdehyde (MDA), nitric oxide (NO), and reduced glutathione (GSH) and the enzymatic activities of sodium-potassium-ATPase (Na+, K+-ATPase), Acetylcholinesterase (AChE), and monoamine oxidase (MAO) and monoamine levels (dopamine (DA), norepinephrine (NE) and serotonin (5-HT) in the midbrain and striatum of reserpine-induced PD in an animal model treated with PBM. Furthermore, the locomotor behavior of the animals has been determined by the open field test. Animals were divided into three groups; the control group, the PD-induced model group, and the PD-induced model treated with the PBM group. Non-invasive treatment of animals for 14 days with 100 mW, 830 nm laser has demonstrated successful attainment in the recovery of oxidative stress, and enzymatic activities impairments induced by reserpine (0.2 mg/kg) in both midbrain and striatum of adult male Wistar rats. PBM also improved the decrease in DA, NE, and 5-HT in the investigated brain regions. On a behavioral level, animals showed improvement in their locomotion activity. These findings have shed more light on some mechanisms underlying the treatment potential of PBM and displayed the safety, easiness, and efficacy of PBM treatment as an alternative to pharmacological treatment for PD.
Collapse
Affiliation(s)
- Haitham S Mohammed
- Faculty of Science, Biophysics Department, Cairo University, Giza, Egypt.
| | - Eman N Hosny
- Medical Division, Medical Physiology Department, National Research Centre, Giza, Egypt
| | - Hussein G Sawie
- Medical Division, Medical Physiology Department, National Research Centre, Giza, Egypt
| | - Yasser A Khadrawy
- Medical Division, Medical Physiology Department, National Research Centre, Giza, Egypt
| |
Collapse
|
25
|
Mahamud N, Songvut P, Muangnoi C, Rodsiri R, Dahlan W, Tansawat R. Untargeted metabolomics reveal pathways associated with neuroprotective effect of oxyresveratrol in SH-SY5Y cells. Sci Rep 2023; 13:20385. [PMID: 37989867 PMCID: PMC10663518 DOI: 10.1038/s41598-023-47558-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 11/15/2023] [Indexed: 11/23/2023] Open
Abstract
Oxyresveratrol has been documented benefits for neurodegenerative disease. However, the specific molecular mechanisms and pathways involved is currently limited. This study aimed to investigate the potential neuroprotective mechanisms of oxyresveratrol using rotenone-induced human neuroblastoma SH-SY5Y cytotoxicity. Cells were divided into the following groups: control, rotenone, and oxyresveratrol pre-treated before being exposed to rotenone. Cellular assays were performed to investigate neuroprotective effects of oxyresveratrol. The results showed that 20 μM oxyresveratrol was effective in preventing rotenone-induced cell death and decreasing ROS levels in the cells. The alteration of metabolites and pathways involved in the neuroprotective activities of oxyresveratrol were further investigated using LC-QTOF-MS/MS untargeted metabolomics approach. We hypothesized that oxyresveratrol's neuroprotective effects would be associated with neurodegenerative pathways. A total of 294 metabolites were identified. 7,8-dihydrobiopterin exhibited the highest VIP scores (VIP > 3.0; p < 0.05), thus considered a biomarker in this study. Our results demonstrated that pretreatment with oxyresveratrol upregulated the level of 7,8-dihydrobiopterin compared to the positive control. Pathway analysis verified that 7,8-dihydrobiopterin was primarily associated with phenylalanine, tyrosine, and tryptophan metabolism (impact = 1, p < 0.001), serving as essential cofactors for enzymatic function in the dopamine biosynthesis pathway. In conclusion, oxyresveratrol may be benefit for the prevention of neurodegenerative diseases by increasing 7,8-dihydrobiopterin concentration.
Collapse
Affiliation(s)
- Nureesun Mahamud
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- The Halal Science Center, Chulalongkorn University, Bangkok, Thailand
| | - Phanit Songvut
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Chawanphat Muangnoi
- Cell and Animal Model Unit, Institute of Nutrition, Mahidol University, Nakhon Pathom, Thailand
| | - Ratchanee Rodsiri
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Winai Dahlan
- The Halal Science Center, Chulalongkorn University, Bangkok, Thailand
| | - Rossarin Tansawat
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand.
- Metabolomics for Life Sciences Research Unit, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
26
|
Tsikas D. GC-MS and GC-MS/MS measurement of malondialdehyde (MDA) in clinical studies: Pre-analytical and clinical considerations. J Mass Spectrom Adv Clin Lab 2023; 30:10-24. [PMID: 37637438 PMCID: PMC10458701 DOI: 10.1016/j.jmsacl.2023.08.001] [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: 01/27/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023] Open
Abstract
Malondialdehyde (MDA; 1,3-propanedial, OHC-CH2-CHO) is one of the most frequently measured biomarkers of oxidative stress in plasma and serum. L-Arginine (Arg) is the substrate of nitric oxide synthases (NOS), which convert L-arginine to nitric oxide (NO) and L-citrulline. The Arg/NO pathway comprises several members, including the endogenous NOS-activity inhibitor asymmetric dimethylarginine (ADMA) and its major metabolite dimethyl amine (DMA), and nitrite and nitrate, the major NO metabolites. Reliable measurement of MDA and members of the Arg/NO pathway in plasma, serum, urine and in other biological samples, such as saliva and cerebrospinal fluid, is highly challenging both for analytical and pre-analytical reasons. In our group, we use validated gas chromatography-mass spectrometry (GC-MS) and gas chromatography-tandem mass spectrometry (GC-MS/MS) methods for the quantitative determination in clinical studies of MDA as a biomarker of oxidative stress, and various Arg/NO metabolites that describe the status of this pathway. Here, the importance of pre-analytical issues, which has emerged from the use of GC-MS and GC-MS/MS in clinico-pharmacological studies, is discussed. Paradigmatically, two studies on the long-term oral administration of L-arginine dihydrochloride to patients suffering from peripheral arterial occlusive disease (PAOD) or coronary artery disease (CAD) were considered. Pre-analytical issues that were addressed include blood sampling, plasma or serum storage, study design (notably in long-term studies), and the alternative of measuring MDA in human urine.
Collapse
Affiliation(s)
- Dimitrios Tsikas
- Hannover Medical School, Institute of Toxicology, Core Unit Proteomics, 30623 Hannover, Germany
| |
Collapse
|
27
|
Siddiqui T, Bhatt LK. Targeting Sigma-1 Receptor: A Promising Strategy in the Treatment of Parkinson's Disease. Neurochem Res 2023; 48:2925-2935. [PMID: 37259012 PMCID: PMC10231286 DOI: 10.1007/s11064-023-03960-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
Parkinson's disease is a neurodegenerative disease affecting mainly the elderly population. It is characterized by the loss of dopaminergic neurons of the substantia nigra pars compacta region. Parkinson's disease patients exhibit motor symptoms like tremors, rigidity, bradykinesia/hypokinesia, and non-motor symptoms like depression, cognitive decline, delusion, and pain. Major pathophysiological factors which contribute to neuron loss include excess/misfolded alpha-synuclein aggregates, microglial cell-mediated neuroinflammation, excitotoxicity, oxidative stress, and defective mitochondrial function. Sigma-1 receptors are molecular chaperones located at mitochondria-associated ER membrane. Their activation (by endogenous ligands or agonists) has shown neuroprotective and neurorestorative effects in various diseases. This review discusses the roles of activated Sig-1 receptors in modulating various pathophysiological features of Parkinson's disease like alpha-synuclein aggregates, neuroinflammation, excitotoxicity, and oxidative stress.
Collapse
Affiliation(s)
- Talha Siddiqui
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India.
| |
Collapse
|
28
|
Adeyeye TA, Babatunde BR, Ehireme SE, Shallie PD. Caffeine alleviates anxiety-like behavior and brainstem lesions in a rotenone-induced rat model of Parkinson's disease. J Chem Neuroanat 2023; 132:102315. [PMID: 37481171 DOI: 10.1016/j.jchemneu.2023.102315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/03/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor and non-motor symptoms. In 2016, approximately 6.1 million individuals were affected by PD, with 211,296 deaths attributed to the disease. The understanding of PD initially came from the observation of dopaminergic system alterations in a specific region of the brainstem, indicating that the core motor and non-motor features of PD are closely associated with brainstem dysfunction. The primary treatment approach for PD revolves around dopamine replacement, as many of the symptoms are responsive to this therapeutic intervention. However, long-term administration of this approach is linked to several complications, and a definitive gold-standard therapy for PD is yet to be identified. The pharmacological management of PD has been challenging and inconsistent, mainly due to the unclear underlying cause of the disease. This study aims to evaluate the effects of caffeine on the brainstem of rats with PD induced by rotenone. METHODOLOGY Fifty adult male Wistar rats weighing between 150 and 200 g were used in this study. The rats were randomly divided into five groups of ten rats each: Vehicle Group, Rotenone-only treated Group (rotenone only treated with 3 mg/kg, intraperitoneal administration [IP]), Preventive Group (caffeine 30 mg/kg + rotenone 3 mg/kg, IP), Curative Group (rotenone 3 mg/kg + caffeine 30 mg/kg, IP), and Caffeine only treated Group (caffeine only treated with 30 mg/kg, IP). The animals underwent neurobehavioral assessments, followed by sacrifice. The brains were then excised, weighed, and processed histologically. Appropriate brain sections were taken and processed. Photomicrographs were obtained, morphometric and statistical analysis was performed using an Omax LED digital RESULTS: The results demonstrated a significant (p < 0.05) reduction in body weight and relative brain weight, which were increased by caffeine treatments. Rotenone administration led to histological changes similar to those observed in PD, including neuronal structural derangement, degenerated nerve fibers, loss of myelinated neurons, and Nissl substance, as well as downregulation in the expressions of NRF2 and TH in the midbrain. However, these pathological features were counteracted or ameliorated by caffeine treatment. CONCLUSION Our study contributes additional evidence to the growing body of research supporting the therapeutic potential of caffeine in Parkinson's disease (PD). The results underscore the neuroprotective properties of caffeine and its capacity to mitigate oxidative stress by modulating TH (tyrosine hydroxylase) and cytoplasmic NRF2 (nuclear factor erythroid 2-related factor 2) in the mesencephalon. These findings suggest that caffeine holds promise as a viable treatment option for PD.
Collapse
|
29
|
Dilnashin H, Birla H, Keswani C, Singh SS, Zahra W, Rathore AS, Singh R, Keshri PK, Singh SP. Neuroprotective Effects of Tinospora cordifolia via Reducing the Oxidative Stress and Mitochondrial Dysfunction against Rotenone-Induced PD Mice. ACS Chem Neurosci 2023; 14:3077-3087. [PMID: 37579290 DOI: 10.1021/acschemneuro.3c00216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023] Open
Abstract
Oxidative stress and mitochondrial dysfunction are leading mechanisms that play a crucial role in the progression of Parkinson's disease (PD). Tinospora cordifolia shows a wide range of biological activities including immunomodulatory, antimicrobial, antioxidant, and anti-inflammatory properties. This study explored the neuroprotective activities of T. cordifolia ethanolic extract (TCE) against Rotenone (ROT)-intoxicated Parkinsonian mice. Four experimental groups of mice were formed: control, ROT (2 mg/kg body wt, subcutaneously), TCE (200 mg/kg body wt, oral) + ROT, and TCE only. Mice were pretreated with TCE for a week and then simultaneously injected with ROT for 35 days. Following ROT-intoxication, motor activities, antioxidative potential, and mitochondrial dysfunction were analyzed. Decrease in the activity of the mitochondrial electron transport chain (mETC) complex, loss of mitochondrial membrane potential (Ψm), increase in Bax/Bcl-2 (B-cell lymphoma 2) ratio, and caspase-3 expression are observed in the ROT-intoxicated mice group. Our results further showed ROT-induced reactive oxygen species (ROS)-mediated alpha-synuclein (α-syn) accumulation and mitochondrial dysfunction. However, pre- and cotreatment with TCE along with ROT-intoxication significantly reduced α-syn aggregation and improved mitochondrial functioning in cells by altering mitochondrial potential and increasing mETC activity. TCE also decreases the Bax/Bcl-2 ratio and also the expression of caspase-3, thus reducing apoptosis of the cell. Altogether, TCE is effective in protecting neurons from rotenone-induced cytotoxicity in the Parkinsonian mouse model by modulating oxidative stress, ultimately reducing mitochondrial dysfunction and cell death.
Collapse
Affiliation(s)
- Hagera Dilnashin
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Hareram Birla
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Chetan Keswani
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Saumitra Sen Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Walia Zahra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Aaina Singh Rathore
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Richa Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Priyanka Kumari Keshri
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Surya Pratap Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, U.P., India
| |
Collapse
|
30
|
Medeiros TB, Cosendey P, Gerin DR, de Sousa GF, Portal TM, Monteiro-de-Barros C. The effect of the sulfation patterns of dermatan and chondroitin sulfate from vertebrates and ascidians on their neuritogenic and neuroprotective properties. Int J Biol Macromol 2023; 247:125830. [PMID: 37454999 DOI: 10.1016/j.ijbiomac.2023.125830] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/08/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Neurodegeneration is caused by the progressive loss of the structure and function of neurons, leading to cell death, and it is the main cause of many neurodegenerative diseases. Many molecules, such as glycosaminoglycans (GAGs), have been studied for their potential to prevent or treat these diseases. They are widespread in nature and perform an important role in neuritogenesis and neuroprotection. Here we investigated the neuritogenic and neuroprotective role of Phallusia nigra dermatan sulfate (PnD2,6S) and compared it with two distinct structures of chondroitin sulfate (C6S) and dermatan sulfate (D4S). For this study, a neuro 2A murine neuroblastoma cell line was used, and a chemical lesion was induced by the pesticide rotenone (ROT). We observed that PnD2,6S + ROT had a better neuritogenic effect than either C6S + ROT or D4S + ROT at a lower concentration (0.05 μg/mL). When evaluating the mitochondrial membrane potential, PnD2,6S showed a neuroprotective effect at a concentration of 0.4 μg/mL. These data indicate different mechanisms underlying this neuronal potential, in which the sulfation pattern is important for neuritogenic activity, while for neuroprotection all DS/CS structures had similar effects. This finding leads to a better understanding the chemical structures of PnD2,6S, C6S, and D4S and their therapeutic potential.
Collapse
Affiliation(s)
- Taiane Barreto Medeiros
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil; Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil
| | - Paloma Cosendey
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil
| | - Diovana Ramos Gerin
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil; Programa de Pós-Graduação em Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, UENF, Av. Alberto Lamego, 2000, Campos dos Goytacazes CEP: 28013-602, RJ, Brazil
| | - Graziele Fonseca de Sousa
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil; Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil
| | - Taynan Motta Portal
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil; Programa de Pós-Graduação em Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, UENF, Av. Alberto Lamego, 2000, Campos dos Goytacazes CEP: 28013-602, RJ, Brazil
| | - Cintia Monteiro-de-Barros
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil; Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil; Programa de Pós-Graduação em Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, UENF, Av. Alberto Lamego, 2000, Campos dos Goytacazes CEP: 28013-602, RJ, Brazil.
| |
Collapse
|
31
|
Qi R, Sammler E, Gonzalez-Hunt CP, Barraza I, Pena N, Rouanet JP, Naaldijk Y, Goodson S, Fuzzati M, Blandini F, Erickson KI, Weinstein AM, Lutz MW, Kwok JB, Halliday GM, Dzamko N, Padmanabhan S, Alcalay RN, Waters C, Hogarth P, Simuni T, Smith D, Marras C, Tonelli F, Alessi DR, West AB, Shiva S, Hilfiker S, Sanders LH. A blood-based marker of mitochondrial DNA damage in Parkinson's disease. Sci Transl Med 2023; 15:eabo1557. [PMID: 37647388 PMCID: PMC11135133 DOI: 10.1126/scitranslmed.abo1557] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/11/2023] [Indexed: 09/01/2023]
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder, and neuroprotective or disease-modifying interventions remain elusive. High-throughput markers aimed at stratifying patients on the basis of shared etiology are required to ensure the success of disease-modifying therapies in clinical trials. Mitochondrial dysfunction plays a prominent role in the pathogenesis of PD. Previously, we found brain region-specific accumulation of mitochondrial DNA (mtDNA) damage in PD neuronal culture and animal models, as well as in human PD postmortem brain tissue. To investigate mtDNA damage as a potential blood-based marker for PD, we describe herein a PCR-based assay (Mito DNADX) that allows for the accurate real-time quantification of mtDNA damage in a scalable platform. We found that mtDNA damage was increased in peripheral blood mononuclear cells derived from patients with idiopathic PD and those harboring the PD-associated leucine-rich repeat kinase 2 (LRRK2) G2019S mutation in comparison with age-matched controls. In addition, mtDNA damage was elevated in non-disease-manifesting LRRK2 mutation carriers, demonstrating that mtDNA damage can occur irrespective of a PD diagnosis. We further established that Lrrk2 G2019S knock-in mice displayed increased mtDNA damage, whereas Lrrk2 knockout mice showed fewer mtDNA lesions in the ventral midbrain, compared with wild-type control mice. Furthermore, a small-molecule kinase inhibitor of LRRK2 mitigated mtDNA damage in a rotenone PD rat midbrain neuron model and in idiopathic PD patient-derived lymphoblastoid cell lines. Quantifying mtDNA damage using the Mito DNADX assay may have utility as a candidate marker of PD and for measuring the pharmacodynamic response to LRRK2 kinase inhibitors.
Collapse
Affiliation(s)
- Rui Qi
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC 27710, USA
| | - Esther Sammler
- Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, DD1 5EH UK
| | - Claudia P. Gonzalez-Hunt
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC 27710, USA
| | - Ivana Barraza
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC 27710, USA
| | - Nicholas Pena
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC 27710, USA
| | - Jeremy P. Rouanet
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yahaira Naaldijk
- Department of Anesthesiology and Department of Physiology, Pharmacology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Steven Goodson
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC 27710, USA
| | - Marie Fuzzati
- IRCCS Mondino Foundation, National Institute of Neurology, Pavia 27100, Italy
| | - Fabio Blandini
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Kirk I. Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- AdventHealth Research Institute, Neuroscience, Orlando, FL 32804, USA
| | - Andrea M. Weinstein
- Department of Psychiatry, School of Medicine, University of Pittsburgh, PA 15213, USA
| | - Michael W. Lutz
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - John B. Kwok
- School of Medical Sciences, Faculty of Medicine and Health and the Brain and Mind Centre, University of Sydney, Camperdown, New South Wales 2050, Australia
| | - Glenda M. Halliday
- School of Medical Sciences, Faculty of Medicine and Health and the Brain and Mind Centre, University of Sydney, Camperdown, New South Wales 2050, Australia
| | - Nicolas Dzamko
- School of Medical Sciences, Faculty of Medicine and Health and the Brain and Mind Centre, University of Sydney, Camperdown, New South Wales 2050, Australia
| | - Shalini Padmanabhan
- Michael J. Fox Foundation for Parkinson’s Research, Grand Central Station, P.O. Box 4777, New York, NY 10120, USA
| | - Roy N. Alcalay
- Columbia University Irving Medical Center, New York, NY 10032, USA
- Movement Disorders Unit, Neurological Institute, Tel Aviv Sourasky Medical Centre, Sackler School of Medicine, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Cheryl Waters
- Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Penelope Hogarth
- Departments of Molecular and Medical Genetics and Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Tanya Simuni
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Danielle Smith
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Connie Marras
- Edmond J. Safra Program in Parkinson’s Disease, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Canada
| | - Francesca Tonelli
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, DD1 5EH UK
| | - Dario R. Alessi
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, DD1 5EH UK
| | - Andrew B. West
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology and Medicine, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Sabine Hilfiker
- Department of Anesthesiology and Department of Physiology, Pharmacology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Laurie H. Sanders
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC 27710, USA
| |
Collapse
|
32
|
Louati K, Kolsi F, Kallel R, Gdoura Y, Borni M, Hakim LS, Zribi R, Choura S, Maalej A, Sayadi S, Chamkha M, Mnif B, Khemakhem Z, Boudawara TS, Boudawara MZ, Safta F. Research of Pesticide Metabolites in Human Brain Tumor Tissues by Chemometrics-Based Gas Chromatography-Mass Spectrometry Analysis for a Hypothetical Correlation between Pesticide Exposure and Risk Factor of Central Nervous System Tumors. ACS OMEGA 2023; 8:29812-29835. [PMID: 37599976 PMCID: PMC10433342 DOI: 10.1021/acsomega.3c04592] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 07/25/2023] [Indexed: 08/22/2023]
Abstract
Pesticides are widely used, resulting in continuing human exposure with potential health impacts. Some exposures related to agricultural works have been associated with neurological disorders. Since the 2000s, the hypothesis of the role of pesticides in the occurrence of central nervous system (CNS) tumors has been better documented in the literature. However, the etiology of childhood brain cancers still remains largely unknown. The major objective of this work was to assess the potential role of pesticide exposure as a risk factor for CNS tumors based on questionnaires and statistical analysis of information collected from patients hospitalized in the Neurosurgery Department of the Habib Bourguiba Hospital Medium in Sfax, Tunisia, during the period from January 1, 2022, to May 31, 2023. It also aimed to develop a simple and rapid analytical method by the gas chromatography-mass spectrometry technique for the research traces of pesticide metabolites in some collected human brain tumor tissues in order to more emphasize our hypothesis for such a correlation between pesticide exposure and brain tumor development. Patients with a history of high-risk exposure were selected to conduct further analysis. Chemometric methods were adapted to discern intrinsic variation between pathological and control groups and ascertain effective separation with the identification of differentially expressed metabolites accountable for such variations. Three samples revealed traces of pesticide metabolites that were mostly detected at an early age. The histopathological diagnosis was medulloblastoma for a 10-year-old child and high-grade gliomas for 27- and 35-year-old adults. The bivariate analyses (odds ratio >1 and P value <5%) confirmed the great probability of developing cancer by an exposure case. The Cox proportional hazards model revealed the risk of carcinogenicity beyond the age of 50 as a long-term effect of pesticide toxicity. Our study supports the correlation between pesticide exposure and the risk of development of human brain tumors, suggesting that preconception pesticide exposure, and possibly exposure during pregnancy, is associated with an increased childhood brain tumor risk. This hypothesis was enhanced in identifying traces of metabolites from the carbamate insecticide class known for their neurotoxicity and others from pyridazinone, organochlorines (OCs), triazole fungicide, and N-nitroso compounds known for their carcinogenicity. The 2D-OXYBLOT analysis confirmed the neurotoxicity effect of insecticides to induce oxidative damage in CNS cells. Aldicarb was implicated in brain carcinogenicity confirmed by the identification of oxime metabolites in a stress degradation study. Revealing "aziridine" metabolites from the OC class may better emphasize the theory of detecting traces of pesticide metabolites at an early age. Overall, our findings lead to the recommendation of limiting the residential use of pesticides and the support of public health policies serving this objective that we need to be vigilant in the postmarketing surveillance of human health impacts.
Collapse
Affiliation(s)
- Kaouthar Louati
- Faculty
of Pharmacy, Laboratory of Pharmacology, Analytics and Galenic Drug
Development, LR12ES09, University of Monastir, Road Avicenne, 5000 Monastir, Tunisia
| | - Fatma Kolsi
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Rim Kallel
- Laboratory
of Pathological Anatomy and Cytology, Habib
Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Yassine Gdoura
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Mahdi Borni
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Leila Sellami Hakim
- Laboratory
of Pathological Anatomy and Cytology, Habib
Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Rania Zribi
- Higher Institute
of Applied Studies to Humanities of Tunis (ISEAHT), University of Tunis, 11 Road of Jebel Lakdhar, 1005 Tunis, Tunisia
| | - Sirine Choura
- Laboratory
of Environmental Bioprocesses, Centre of
Biotechnology of Sfax, Road of Sidi-Mansour, P.O. Box 1177, 3018 Sfax, Tunisia
| | - Amina Maalej
- Laboratory
of Environmental Bioprocesses, Centre of
Biotechnology of Sfax, Road of Sidi-Mansour, P.O. Box 1177, 3018 Sfax, Tunisia
| | - Sami Sayadi
- Biotechnology
Program, Center for Sustainable Development, College of Arts and Sciences, Qatar University, 2713 Doha, Qatar
| | - Mohamed Chamkha
- Laboratory
of Environmental Bioprocesses, Centre of
Biotechnology of Sfax, Road of Sidi-Mansour, P.O. Box 1177, 3018 Sfax, Tunisia
| | - Basma Mnif
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Zouheir Khemakhem
- Legal
Medicine Department, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Tahya Sellami Boudawara
- Laboratory
of Pathological Anatomy and Cytology, Habib
Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Mohamed Zaher Boudawara
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Fathi Safta
- Faculty
of Pharmacy, Laboratory of Pharmacology, Analytics and Galenic Drug
Development, LR12ES09, University of Monastir, Road Avicenne, 5000 Monastir, Tunisia
| |
Collapse
|
33
|
Fagen SJ, Burgess JD, Lim MJ, Amerna D, Kaya ZB, Faroqi AH, Perisetla P, DeMeo NN, Stojkovska I, Quiriconi DJ, Mazzulli JR, Delenclos M, Boschen SL, McLean PJ. Honokiol decreases alpha-synuclein mRNA levels and reveals novel targets for modulating alpha-synuclein expression. Front Aging Neurosci 2023; 15:1179086. [PMID: 37637959 PMCID: PMC10449643 DOI: 10.3389/fnagi.2023.1179086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
Background Intracytoplasmic inclusions comprised of aggregated alpha-synuclein (αsyn) represent a key histopathological feature of neurological disorders collectively termed "synucleinopathies," which includes Parkinson's disease (PD). Mutations and multiplications in the SNCA gene encoding αsyn cause familial forms of PD and a large body of evidence indicate a correlation between αsyn accumulation and disease. Decreasing αsyn expression is recognized as a valid target for PD therapeutics, with down-regulation of SNCA expression potentially attenuating downstream cascades of pathologic events. Here, we evaluated if Honokiol (HKL), a polyphenolic compound derived from magnolia tree bark with demonstrated neuroprotective properties, can modulate αsyn levels in multiple experimental models. Methods Human neuroglioma cells stably overexpressing αsyn, mouse primary neurons, and human iPSC-derived neurons were exposed to HKL and αsyn protein and SNCA messenger RNA levels were assessed. The effect of HKL on rotenone-induced overexpression of αsyn levels was further assessed and transcriptional profiling of mouse cortical neurons treated with HKL was performed to identify potential targets of HKL. Results We demonstrate that HKL can successfully reduce αsyn protein levels and SNCA expression in multiple in vitro models of PD with our data supporting a mechanism whereby HKL acts by post-transcriptional modulation of SNCA rather than modulating αsyn protein degradation. Transcriptional profiling of mouse cortical neurons treated with HKL identifies several differentially expressed genes (DEG) as potential targets to modulate SNCA expression. Conclusion This study supports a HKL-mediated downregulation of SNCA as a viable strategy to modify disease progression in PD and other synucleinopathies. HKL has potential as a powerful tool for investigating SNCA gene modulation and its downstream effects.
Collapse
Affiliation(s)
- Sara J. Fagen
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Jeremy D. Burgess
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Melina J. Lim
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Danilyn Amerna
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Zeynep B. Kaya
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Ayman H. Faroqi
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Priyanka Perisetla
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Natasha N. DeMeo
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Iva Stojkovska
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Drew J. Quiriconi
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Joseph R. Mazzulli
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Marion Delenclos
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Suelen L. Boschen
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Rochester, MN, United States
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, United States
| | - Pamela J. McLean
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Rochester, MN, United States
| |
Collapse
|
34
|
Zhang W, Chen S, Huang X, Tong H, Niu H, Lu L. Neuroprotective effect of a medium-chain triglyceride ketogenic diet on MPTP-induced Parkinson's disease mice: a combination of transcriptomics and metabolomics in the substantia nigra and fecal microbiome. Cell Death Discov 2023; 9:251. [PMID: 37460539 DOI: 10.1038/s41420-023-01549-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/18/2023] [Accepted: 07/05/2023] [Indexed: 07/20/2023] Open
Abstract
The ketogenic diet (KD) is a low carbohydrate and high-fat protein diet. It plays a protective role in neurodegenerative diseases by elevating the levels of ketone bodies in blood, regulating central and peripheral metabolism and mitochondrial functions, inhibiting neuroinflammation and oxidative stress, and altering the gut microbiota. However, studies on ketogenic therapy for Parkinson's disease (PD) are still in their infancy. Therefore, we examined the possible protective effect of KD in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model, examined the mouse gut microbiota and its metabolites, and performed transcriptomics and metabolomics on the substantia nigra of mice. Our results showed that a long-term medium-chain triglyceride KD (MCT-KD) significantly reduced MPTP-induced damage to dopaminergic (DA) neurons, exerted antioxidant stress through the PI3K/Akt/Nrf2 pathway, and reversed oxidative stress in DA neurons. The MCT-KD also reduced mitochondrial loss, promoted ATP production, and inhibited the activation of microglia to protect DA neurons in MPTP-induced PD mice. MCT-KD altered the gut microbiota and consequently changed the metabolism of substantia nigra neurons through gut microbiota metabolites. Compared to the MPTP group, MCT-KD increased the abundance of gut microbiota, including Blautia and Romboutsia. MCT-KD also affects purine metabolism in the substantia nigra pars compacta (SNpc) by altering fecal metabolites. This study shows that MCT-KD has multiple protective effects against PD.
Collapse
Affiliation(s)
- Wenlong Zhang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510120, China
| | - Shiyu Chen
- Department of General practice, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510282, China
| | - Xingting Huang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510120, China
| | - Huichun Tong
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, 510632, China
| | - Hongxin Niu
- General practice and Special medical service center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510282, China.
| | - Lingli Lu
- Department of General practice, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510282, China.
| |
Collapse
|
35
|
Romero-Sanz S, Caldero-Escudero E, Álvarez-Illera P, Santo-Domingo J, Fonteriz RI, Montero M, Álvarez J. SERCA inhibition improves lifespan and healthspan in a chemical model of Parkinson disease in Caenorhabditis elegans. Front Pharmacol 2023; 14:1182428. [PMID: 37284303 PMCID: PMC10239880 DOI: 10.3389/fphar.2023.1182428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/09/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction: The high prevalence of neurodegenerative diseases in our population and the lack of effective treatments encourage the search for new therapeutic targets for these pathologies. We have recently described that submaximal inhibition of the Sarco-Endoplasmic Reticulum Ca2+ ATPase (SERCA), the main responsible for ER calcium storage, is able to increase lifespan in Caenorhabditis elegans worms by mechanisms involving mitochondrial metabolism and nutrient-sensitive pathways. Methods: We have studied here the effects of submaximal SERCA inhibition in a chemical model of Parkinson's disease (PD) induced in C. elegans worms by treatment with the mitochondrial complex I inhibitor rotenone. For specific SERCA inhibition, we treated worms with RNAi against sca-1, the sole orthologue of SERCA in C. elegans. Results and Discussion: Our results show that rotenone produces alterations in worms that include decreased lifespan, smaller size, reduced fertility, decreased motility, defecation and pumping rate, increased mitochondrial ROS production, reduced mitochondrial membrane potential and oxygen consumption rate, altered mitochondrial structure, and altered ethanol preference in behavioral studies. Most of these alterations were either fully or partially reversed in worms treated with sca-1 RNAi, suggesting that SERCA inhibition could be a novel pharmacological target in the prevention or treatment of neurodegeneration.
Collapse
|
36
|
Geng Y, Wang Z, Zhou J, Zhu M, Liu J, James TD. Recent progress in the development of fluorescent probes for imaging pathological oxidative stress. Chem Soc Rev 2023. [PMID: 37190785 DOI: 10.1039/d2cs00172a] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Oxidative stress is closely related to the physiopathology of numerous diseases. Reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive sulfur species (RSS) are direct participants and important biomarkers of oxidative stress. A comprehensive understanding of their changes can help us evaluate disease pathogenesis and progression and facilitate early diagnosis and drug development. In recent years, fluorescent probes have been developed for real-time monitoring of ROS, RNS and RSS levels in vitro and in vivo. In this review, conventional design strategies of fluorescent probes for ROS, RNS, and RSS detection are discussed from three aspects: fluorophores, linkers, and recognition groups. We introduce representative fluorescent probes for ROS, RNS, and RSS detection in cells, physiological/pathological processes (e.g., Inflammation, Drug Induced Organ Injury and Ischemia/Reperfusion Injury etc.), and specific diseases (e.g., neurodegenerative diseases, epilepsy, depression, diabetes and cancer, etc.). We then highlight the achievements, current challenges, and prospects for fluorescent probes in the pathophysiology of oxidative stress-related diseases.
Collapse
Affiliation(s)
- Yujie Geng
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Zhuo Wang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Jiaying Zhou
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Mingguang Zhu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Jiang Liu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Tony D James
- Department of Chemistry, University of Bath, Bath BA2 7AY, UK.
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
37
|
Lucchi C, Codeluppi A, Filaferro M, Vitale G, Rustichelli C, Avallone R, Mandrioli J, Biagini G. Human Microglia Synthesize Neurosteroids to Cope with Rotenone-Induced Oxidative Stress. Antioxidants (Basel) 2023; 12:antiox12040963. [PMID: 37107338 PMCID: PMC10135967 DOI: 10.3390/antiox12040963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/05/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
We obtained evidence that mouse BV2 microglia synthesize neurosteroids dynamically to modify neurosteroid levels in response to oxidative damage caused by rotenone. Here, we evaluated whether neurosteroids could be produced and altered in response to rotenone by the human microglial clone 3 (HMC3) cell line. To this aim, HMC3 cultures were exposed to rotenone (100 nM) and neurosteroids were measured in the culture medium by liquid chromatography with tandem mass spectrometry. Microglia reactivity was evaluated by measuring interleukin 6 (IL-6) levels, whereas cell viability was monitored by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. After 24 h (h), rotenone increased IL-6 and reactive oxygen species levels by approximately +37% over the baseline, without affecting cell viability; however, microglia viability was significantly reduced at 48 h (p < 0.01). These changes were accompanied by the downregulation of several neurosteroids, including pregnenolone, pregnenolone sulfate, 5α-dihydroprogesterone, and pregnanolone, except for allopregnanolone, which instead was remarkably increased (p < 0.05). Interestingly, treatment with exogenous allopregnanolone (1 nM) efficiently prevented the reduction in HMC3 cell viability. In conclusion, this is the first evidence that human microglia can produce allopregnanolone and that this neurosteroid is increasingly released in response to oxidative stress, to tentatively support the microglia's survival.
Collapse
Affiliation(s)
- Chiara Lucchi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Alessandro Codeluppi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Monica Filaferro
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giovanni Vitale
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Cecilia Rustichelli
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Rossella Avallone
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Jessica Mandrioli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Department of Neurosciences, Ospedale Civile di Baggiovara, Azienda Ospedaliero-Universitaria di Modena, 41126 Modena, Italy
| | - Giuseppe Biagini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
38
|
Di Maio R, Keeney MT, Cechova V, Mortimer A, Sekandari A, Rowart P, Greenamyre JT, Freeman BA, Fazzari M. Neuroprotective actions of a fatty acid nitroalkene in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:55. [PMID: 37029127 PMCID: PMC10082007 DOI: 10.1038/s41531-023-00502-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 03/23/2023] [Indexed: 04/09/2023] Open
Abstract
To date there are no therapeutic strategies that limit the progression of Parkinson's disease (PD). The mechanisms underlying PD-related nigrostriatal neurodegeneration remain incompletely understood, with multiple factors modulating the course of PD pathogenesis. This includes Nrf2-dependent gene expression, oxidative stress, α-synuclein pathology, mitochondrial dysfunction, and neuroinflammation. In vitro and sub-acute in vivo rotenone rat models of PD were used to evaluate the neuroprotective potential of a clinically-safe, multi-target metabolic and inflammatory modulator, the electrophilic fatty acid nitroalkene 10-nitro-oleic acid (10-NO2-OA). In N27-A dopaminergic cells and in the substantia nigra pars compacta of rats, 10-NO2-OA activated Nrf2-regulated gene expression and inhibited NOX2 and LRRK2 hyperactivation, oxidative stress, microglial activation, α-synuclein modification, and downstream mitochondrial import impairment. These data reveal broad neuroprotective actions of 10-NO2-OA in a sub-acute model of PD and motivate more chronic studies in rodents and primates.
Collapse
Affiliation(s)
- Roberto Di Maio
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA.
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Matthew T Keeney
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15261, USA
| | - Veronika Cechova
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15261, USA
| | - Amanda Mortimer
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ahssan Sekandari
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15261, USA
| | - Pascal Rowart
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15261, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15261, USA
| | - Marco Fazzari
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
39
|
Wang Z, Gao C, Zhang L, Sui R. Hesperidin methylchalcone (HMC) hinders amyloid-β induced Alzheimer's disease by attenuating cholinesterase activity, macromolecular damages, oxidative stress and apoptosis via regulating NF-κB and Nrf2/HO-1 pathways. Int J Biol Macromol 2023; 233:123169. [PMID: 36623626 DOI: 10.1016/j.ijbiomac.2023.123169] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/04/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023]
Abstract
Phytocompounds therapy has recently emerged as an effective strategy to treat Alzheimer's disease. Herein, the protective effect of hesperidin methylchalcone (HMC) was evaluated through Alzheimer's disease models of Neuro-2a cells and Wistar rats. The in vitro results showed that HMC possesses significant ability to inhibit the acetylcholinesterase enzyme and exhibiting anti-aggregation and disaggregation properties. Furthermore, HMC could protect the Neuro-2a cells against Aβ-induced neurotoxicity. Simultaneously, HMC treatment significantly improved the cognitive deficits caused by Aβ-peptide on spatial memory in Wistar rats. HMC significantly enhanced the cholinergic effects by inhibiting AChE, BuChE, β-secretase activity, caspase-3 activity, and attenuating macromolecular damages and apoptosis. Notably, HMC reduced the Aβ-induced oxidative stress by activating the antioxidative defence enzymes. In addition, the HMC treatment suppressed the expression of immunocytokines such as p-NF-κB p65, p-IκBα, induced by Aβ; whereas upregulating Nrf2, HO-1 in brain homogenate. These results suggest that HMC could attenuate Aβ-induced neuroinflammation in brain via suppressing NF-κB signalling pathway and activating the Nrf2/HO-1 pathway, thereby improving memory and cognitive impairments in Wistar rats. Overall, the present study reports that HMC can act as a potent candidate with multi-faceted neuroprotective potential against Aβ-induced memory dysfunction in Wistar rats for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Zhuo Wang
- School of Nursing, Jinzhou Medical University, Jinzhou 121099, China
| | - Chao Gao
- School of Nursing, Jinzhou Medical University, Jinzhou 121099, China
| | - Lei Zhang
- School of Nursing, Jinzhou Medical University, Jinzhou 121099, China
| | - Rubo Sui
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121099, China.
| |
Collapse
|
40
|
Wang ZX, Li YL, Pu JL, Zhang BR. DNA Damage-Mediated Neurotoxicity in Parkinson’s Disease. Int J Mol Sci 2023; 24:ijms24076313. [PMID: 37047285 PMCID: PMC10093980 DOI: 10.3390/ijms24076313] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease around the world; however, its pathogenesis remains unclear so far. Recent advances have shown that DNA damage and repair deficiency play an important role in the pathophysiology of PD. There is growing evidence suggesting that DNA damage is involved in the propagation of cellular damage in PD, leading to neuropathology under different conditions. Here, we reviewed the current work on DNA damage repair in PD. First, we outlined the evidence and causes of DNA damage in PD. Second, we described the potential pathways by which DNA damage mediates neurotoxicity in PD and discussed the precise mechanisms that drive these processes by DNA damage. In addition, we looked ahead to the potential interventions targeting DNA damage and repair. Finally, based on the current status of research, key problems that need to be addressed in future research were proposed.
Collapse
Affiliation(s)
| | | | - Jia-Li Pu
- Correspondence: (J.-L.P.); (B.-R.Z.); Tel./Fax: +86-571-87784752 (J.-L.P. & B.-R.Z.)
| | - Bao-Rong Zhang
- Correspondence: (J.-L.P.); (B.-R.Z.); Tel./Fax: +86-571-87784752 (J.-L.P. & B.-R.Z.)
| |
Collapse
|
41
|
Moradi Vastegani S, Nasrolahi A, Ghaderi S, Belali R, Rashno M, Farzaneh M, Khoshnam SE. Mitochondrial Dysfunction and Parkinson's Disease: Pathogenesis and Therapeutic Strategies. Neurochem Res 2023:10.1007/s11064-023-03904-0. [PMID: 36943668 DOI: 10.1007/s11064-023-03904-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/23/2023]
Abstract
Parkinson's disease (PD) is a common age-related neurodegenerative disorder whose pathogenesis is not completely understood. Mitochondrial dysfunction and increased oxidative stress have been considered as major causes and central events responsible for the progressive degeneration of dopaminergic (DA) neurons in PD. Therefore, investigating mitochondrial disorders plays a role in understanding the pathogenesis of PD and can be an important therapeutic target for this disease. This study discusses the effect of environmental, genetic and biological factors on mitochondrial dysfunction and also focuses on the mitochondrial molecular mechanisms underlying neurodegeneration, and its possible therapeutic targets in PD, including reactive oxygen species generation, calcium overload, inflammasome activation, apoptosis, mitophagy, mitochondrial biogenesis, and mitochondrial dynamics. Other potential therapeutic strategies such as mitochondrial transfer/transplantation, targeting microRNAs, using stem cells, photobiomodulation, diet, and exercise were also discussed in this review, which may provide valuable insights into clinical aspects. A better understanding of the roles of mitochondria in the pathophysiology of PD may provide a rationale for designing novel therapeutic interventions in our fight against PD.
Collapse
Affiliation(s)
- Sadegh Moradi Vastegani
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Imam Khomeini Hospital Clinical Research Development Unit, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shahab Ghaderi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rafie Belali
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masome Rashno
- Asadabad School of Medical Sciences, Asadabad, Iran
- Student Research Committee, Asadabad School of Medical Sciences, Asadabad, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
42
|
Roy T, Chatterjee A, Swarnakar S. Rotenone induced neurodegeneration is mediated via cytoskeleton degradation and necroptosis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119417. [PMID: 36581087 DOI: 10.1016/j.bbamcr.2022.119417] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/07/2022] [Accepted: 12/16/2022] [Indexed: 12/28/2022]
Abstract
Rotenone has widespread beneficial effects in agriculture, fisheries and animal husbandries; however prolonged exposure causes a detrimental effect on the health of personnel working in such industries. Rotenone during its extraction, formulation or usage may cross the blood brain barrier leading to neurodegeneration and the development of Parkinson's disease like symptoms. It is a known inhibitor of the mitochondrial ETC complex I and responsible for impairing the OXPHOS system. Our study showed that rotenone exposure results in an increased production of ROS and decreased ATP level along with a conspicuous loss of mitochondrial membrane potential in N2A cells. The transcription and expression pattern of cofilin, a key component of actin cytoskeleton, was also altered after rotenone exposure; leading to the actin cytoskeleton degradation. We further observed an increased expression, as well as activity of matrix metalloproteinase9 (MMP9) in rotenone exposed N2A cells; suggesting the involvement of inflammation upon rotenone exposure. Simultaneously, an opposite pattern was noticed for the tissue inhibitors of metalloproteinases-1 (TIMP-1) protein, which is a known modulator of MMP9 activity. Additionally, the localization of MMP9 along with alpha-synuclein, UCHL1 and cofilin suggested their close proximity and cross interaction upon rotenone treatment. Furthermore, we observed significant increase in the level of TNF-α upon rotenone exposure along with the phosphorylation of RIPK1, RIPK3 and MLKL that has been identified as the necroptosis markers leading to programmed necroptotic death.
Collapse
Affiliation(s)
- Tapasi Roy
- CSIR-Indian Institute of Chemical Biology, Infectious Diseases and Immunology Division, Kolkata, West Bengal, India
| | - Abhishek Chatterjee
- CSIR-Indian Institute of Chemical Biology, Infectious Diseases and Immunology Division, Kolkata, West Bengal, India
| | - Snehasikta Swarnakar
- CSIR-Indian Institute of Chemical Biology, Infectious Diseases and Immunology Division, Kolkata, West Bengal, India.
| |
Collapse
|
43
|
Rasheed MZ, Khatoon R, Talat F, Alam MM, Tabassum H, Parvez S. Melatonin Mitigates Rotenone-Induced Oxidative Stress and Mitochondrial Dysfunction in the Drosophila melanogaster Model of Parkinson's Disease-like Symptoms. ACS OMEGA 2023; 8:7279-7288. [PMID: 36872990 PMCID: PMC9979363 DOI: 10.1021/acsomega.2c03992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 09/12/2022] [Indexed: 06/18/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder; however, its etiology remains elusive. Antioxidants are considered to be a promising approach for decelerating neurodegenerative disease progression owing to extensive examination of the relationship between oxidative stress and neurodegenerative diseases. In this study, we investigated the therapeutic effect of melatonin against rotenone-induced toxicity in the Drosophila model of PD. The 3-5 day old flies were divided into four groups: control, melatonin alone, melatonin and rotenone, and rotenone alone groups. According to their respective groups, flies were exposed to a diet containing rotenone and melatonin for 7 days. We found that melatonin significantly reduced the mortality and climbing ability of Drosophila because of its antioxidative potency. It alleviated the expression of Bcl 2, tyrosine hydroxylase (TH), NADH dehydrogenase, mitochondrial membrane potential, and mitochondrial bioenergetics and decreased caspase 3 expression in the Drosophila model of rotenone-induced PD-like symptoms. These results indicate the neuromodulatory effect of melatonin, and that it is likely modulated against rotenone-induced neurotoxicity by suppressing oxidative stress and mitochondrial dysfunctions.
Collapse
Affiliation(s)
- Md. Zeeshan Rasheed
- Department
of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Rehana Khatoon
- Department
of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Faizia Talat
- Department
of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Mumtaz Alam
- Drug
Design and Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry,
School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Heena Tabassum
- Division
of Basic Medical Sciences, Indian Council
of Medical Research, Ministry of Health and Family Welfare, Govt.
of India, V. Ramalingaswami Bhawan, P.O. Box No. 4911, New Delhi 110029, India
| | - Suhel Parvez
- Department
of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
44
|
Shu DY, Chaudhary S, Cho KS, Lennikov A, Miller WP, Thorn DC, Yang M, McKay TB. Role of Oxidative Stress in Ocular Diseases: A Balancing Act. Metabolites 2023; 13:187. [PMID: 36837806 PMCID: PMC9960073 DOI: 10.3390/metabo13020187] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Redox homeostasis is a delicate balancing act of maintaining appropriate levels of antioxidant defense mechanisms and reactive oxidizing oxygen and nitrogen species. Any disruption of this balance leads to oxidative stress, which is a key pathogenic factor in several ocular diseases. In this review, we present the current evidence for oxidative stress and mitochondrial dysfunction in conditions affecting both the anterior segment (e.g., dry eye disease, keratoconus, cataract) and posterior segment (age-related macular degeneration, proliferative vitreoretinopathy, diabetic retinopathy, glaucoma) of the human eye. We posit that further development of therapeutic interventions to promote pro-regenerative responses and maintenance of the redox balance may delay or prevent the progression of these major ocular pathologies. Continued efforts in this field will not only yield a better understanding of the molecular mechanisms underlying the pathogenesis of ocular diseases but also enable the identification of novel druggable redox targets and antioxidant therapies.
Collapse
Affiliation(s)
- Daisy Y. Shu
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Suman Chaudhary
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Kin-Sang Cho
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Anton Lennikov
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - William P. Miller
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - David C. Thorn
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Menglu Yang
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Tina B. McKay
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
45
|
Shin S, Hwang SK, Mun JY. Changes of lysosome by L-serine in rotenone-treated hippocampal neurons. Appl Microsc 2023; 53:1. [PMID: 36626017 PMCID: PMC9832202 DOI: 10.1186/s42649-022-00084-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/18/2022] [Indexed: 01/11/2023] Open
Abstract
Oxidative stress destroys cellular organelles and damages DNA, eventually leading to degenerative brain disorders. Persistent mitochondrial damage by oxidative stress eventually causes cells to inhibit the function of lysosomes. Rotenone used in this study inhibits complex 1 of the mitochondrial electron transport chain. Due to this inhibition, the production of free radicals is promoted, and oxidative stress can occur. To test as a role of antioxidant, L-serine was treated before treatment of rotenone to HT22 hippocampal cells. Then, changes in the activity and structure of lysosomes were analyzed. As a result, the oxidative stress caused by rotenone in HT22 cells was protected by L-serine. L-serine reduced free radicals in cells, and the damaged lysosomal structure and lysosome activity were also protected.
Collapse
Affiliation(s)
- Sumin Shin
- grid.452628.f0000 0004 5905 0571Neural Circuit Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Su-Kyeong Hwang
- grid.258803.40000 0001 0661 1556Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu, 41944 South Korea ,Astrogen Inc., 440, Hyeoksin-daero, Dong-gu, Daegu, 41072 South Korea
| | - Ji Young Mun
- grid.452628.f0000 0004 5905 0571Neural Circuit Research Group, Korea Brain Research Institute, Daegu, South Korea
| |
Collapse
|
46
|
Pakmanesh F, Mahjoub S, Neamati N, Moslemi D. Chromium and lead levels and alteration in DDPH inhibition in patients with breast cancer undergoing chemotherapy. CASPIAN JOURNAL OF INTERNAL MEDICINE 2023; 14:553-559. [PMID: 37520864 PMCID: PMC10379788 DOI: 10.22088/cjim.14.3.533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 08/01/2023]
Abstract
Background Recently the carcinogenic and toxic effects of some heavy metals such as chromium (Cr), and lead (Pb) through the mechanism of oxidative stress have been reported. Due to the various consequences of chemotherapeutic treatments on body hemostasis, the present study aimed to evaluate the effect of Adriamycin 60 mg/m2 and Cytoxan 600 mg/m2 (AC) chemotherapy on the serum levels of Cr, Pb, and the percent α-diphenyl-β-picrylhydrazyl (DPPH) inhibition. Methods This study was performed on 50 patients with breast cancer at two separate sampling times, the first at the initiation of chemotherapy and the last at the end of three courses of the AC chemotherapy treatment. Serum levels of Cr and Pb were measured using atomic absorption spectrophotometry. The percent DPPH inhibition (% I) and also the effect of age and stage of the disease on the mentioned variables were evaluated. Statistical comparison of the obtained results before and after chemotherapy was performed using paired sample t-test. Intra-group evaluation of age and disease stages was done using an independent sample t-test. Results A significant decrease was observed in the percent DPPH inhibition after 3 courses of chemotherapy (p<0.001). Cr and also Pb were significantly higher in patients with breast cancer after AC chemotherapy (p<0.001). Conclusion According to the results, AC chemotherapy in patients with breast cancer is associated with higher levels of Cr and Pb, which can eventually lead to worsened oxidative stress status in affected patients. However, it seems that these changes do not necessarily depend on age and the stage of the disease.
Collapse
Affiliation(s)
- Fatemeh Pakmanesh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Soleiman Mahjoub
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Nahid Neamati
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Daryush Moslemi
- Cancer Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
47
|
Hong X, Hu Y, Yuan Z, Fang Z, Zhang X, Yuan Y, Guo C. Oxidatively Damaged Nucleic Acid: Linking Diabetes and Cancer. Antioxid Redox Signal 2022; 37:1153-1167. [PMID: 35946074 DOI: 10.1089/ars.2022.0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Our current knowledge of the mechanism between diabetes and cancer is limited. Oxidatively damaged nucleic acid is considered a critical factor to explore the connections between these two diseases. Recent Advances: The link between diabetes mellitus and cancer has attracted increasing attention in recent years. Emerging evidence supports that oxidatively damaged nucleic acid caused by an imbalance between reactive oxygen species generation and elimination is a bridge connecting diabetes and cancer. 8-Oxo-7,8-dihydro-2'-deoxyguanosine and 8-oxo-7,8-dihydroguanosine assume important roles as biomarkers in assessing the relationship between oxidatively damaged nucleic acid and cancer. Critical Issues: The consequences of diabetes are extensive and may lead to the occurrence of cancer by influencing a combination of factors. At present, there is no direct evidence that diabetes causes cancer by affecting a single factor. Furthermore, the difficulty in controlling variables and differences in detection methods lead to poor reliability and repeatability of results, and there are no clear cutoff values for biomarkers to indicate cancer risk. Future Directions: A better understanding of connections as well as mechanisms between diabetes and cancer is still needed. Both diabetes and cancer are currently intractable diseases. Further exploration of the specific mechanism of oxidatively damaged nucleic acid in the connection between diabetes and cancer is urgently needed. In the future, it is necessary to further take oxidatively damaged nucleic acid as an entry point to provide new ideas for the diagnosis and treatment of diabetes and cancer. Experimental drugs targeting the repair process of oxidatively generated damage require an extensive preclinical evaluation and could ultimately provide new treatment strategies for these diseases. Antioxid. Redox Signal. 37, 1153-1167.
Collapse
Affiliation(s)
- Xiujuan Hong
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiqiu Hu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijun Yuan
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihao Fang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxiao Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Yuan
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| | - Cheng Guo
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
48
|
Toki E, Goto S, Setoguchi S, Terada K, Watase D, Yamakawa H, Yamada A, Koga M, Kubota K, Iwasaki K, Karube Y, Matsunaga K, Takata J. Delivery of the reduced form of vitamin K 2(20) to NIH/3T3 cells partially protects against rotenone induced cell death. Sci Rep 2022; 12:19878. [PMID: 36400879 PMCID: PMC9674836 DOI: 10.1038/s41598-022-24456-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Mitochondria generate energy through the action of the electron transport chain (ETC) and ATP synthase. Mitochondrial malfunction can lead to various disorders, including neurodegenerative diseases. Several reports have shown that menaquinone-4 (MK-4, vitamin K2(20)), a safe drug for osteoporosis, may improve mitochondrial function. Here, we hypothesized that the efficient delivery of menahydroquinone-4 (MKH), an active form of MK-4, could exert a supporting effect. We verified the effects of MKH delivery on mitochondrial dysfunction by using MK-4 and MKH ester derivatives in NIH/3T3 mouse fibroblast cells treated with mitochondrial inhibitors. MK-4 and MKH derivatives suppressed cell death, the decline in mitochondrial membrane potential (MMP), excessive reactive oxygen species (ROS) production, and a decrease in intrinsic coenzyme Q9 (CoQ9) induced by rotenone (ROT, complex I inhibitor). MK-4 and MKH derivatives delivered MKH to NIH/3T3 cells, acting as an effective MKH prodrug, proving that the delivered MKH may reflect the mitigation effects on ROT-induced mitochondrial dysfunction. MKH prodrugs are also effective against 3-nitropropionic acid (3-NP, complex II inhibitor) and carbonyl cyanide-m-chlorophenylhydrazone (CCCP, uncoupler)-induced cell death. In conclusion, MKH delivery may mitigate mitochondrial dysfunction by maintaining MMP, ROS, and CoQ9, indicating that MKH prodrugs may be good candidates for treating mitochondrial disorders.
Collapse
Affiliation(s)
- Erina Toki
- grid.411497.e0000 0001 0672 2176Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180 Japan
| | - Shotaro Goto
- grid.411497.e0000 0001 0672 2176Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180 Japan
| | - Shuichi Setoguchi
- grid.411497.e0000 0001 0672 2176Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180 Japan
| | - Kazuki Terada
- grid.412142.00000 0000 8894 6108Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, Himeji, 670-8524 Japan
| | - Daisuke Watase
- grid.411497.e0000 0001 0672 2176Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180 Japan
| | - Hirofumi Yamakawa
- grid.411497.e0000 0001 0672 2176Radioisotope Center, Fukuoka University, Fukuoka, 814-0180 Japan
| | - Ayano Yamada
- grid.411497.e0000 0001 0672 2176Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180 Japan
| | - Mitsuhisa Koga
- grid.411497.e0000 0001 0672 2176Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180 Japan
| | - Kaori Kubota
- grid.411497.e0000 0001 0672 2176Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180 Japan
| | - Katsunori Iwasaki
- grid.411497.e0000 0001 0672 2176Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180 Japan
| | - Yoshiharu Karube
- grid.411497.e0000 0001 0672 2176Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180 Japan
| | - Kazuhisa Matsunaga
- grid.411497.e0000 0001 0672 2176Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180 Japan
| | - Jiro Takata
- grid.411497.e0000 0001 0672 2176Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180 Japan
| |
Collapse
|
49
|
Mirzaei H, Sedighi S, Kouchaki E, Barati E, Dadgostar E, Aschner M, Tamtaji OR. Probiotics and the Treatment of Parkinson's Disease: An Update. Cell Mol Neurobiol 2022; 42:2449-2457. [PMID: 34283340 PMCID: PMC8770674 DOI: 10.1007/s10571-021-01128-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is a progressive neurological disorder characterized by motor and non-motor features. Although some progress has been made in conventional PD treatments, these breakthroughs have yet to show high efficacy in treating this neurodegenerative disease. Probiotics are live microorganisms that confer health benefits on the host when administered in adequate amounts. Probiotics have putative anticancer, antioxidative, anti-inflammatory, and neuroprotective effects. Multiple lines of evidence show that probiotics control and improve several motor and non-motor symptoms in patients and experimental animal models of PD. Probiotic supplementation mediates these pharmacological effects by targeting a variety of cellular and molecular processes, i.e., oxidative stress, inflammatory and anti-inflammatory pathways, as well as apoptosis. Herein, we summarize the effects of probiotics on motor and non-motor symptoms as well as various cellular and molecular pathways in PD.
Collapse
Affiliation(s)
- Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Ebrahim Kouchaki
- Department of Neurology, School of Medicine, Kashan University of Medical Sciences, Kashan, I.R. of Iran
| | - Erfaneh Barati
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, I.R. of Iran
| | - Ehsan Dadgostar
- Department of Psychiatry, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Omid Reza Tamtaji
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
50
|
Hillmann KB, Goethel ME, Erickson NA, Niehaus TD. Identification of a S-(2-succino)cysteine breakdown pathway that uses a novel S-(2-succino) lyase. J Biol Chem 2022; 298:102639. [PMID: 36309089 PMCID: PMC9706529 DOI: 10.1016/j.jbc.2022.102639] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022] Open
Abstract
Succination is the spontaneous reaction between the respiratory intermediate fumarate and cellular thiols that forms stable S-(2-succino)-adducts such as S-(2-succino)cysteine (2SC). 2SC is a biomarker for conditions associated with elevated fumarate levels, including diabetes, obesity, and certain cancers, and succination likely contributes to disease progression. Bacillus subtilis has a yxe operon-encoded breakdown pathway for 2SC that involves three distinct enzymatic conversions. The first step is N-acetylation of 2SC by YxeL to form N-acetyl-2SC (2SNAC). YxeK catalyzes the oxygenation of 2SNAC, resulting in its breakdown to oxaloacetate and N-acetylcysteine, which is deacetylated by YxeP to give cysteine. The monooxygenase YxeK is key to the pathway but is rare, with close homologs occurring infrequently in prokaryote and fungal genomes. The existence of additional 2SC breakdown pathways was not known prior to this study. Here, we used comparative genomics to identify a S-(2-succino) lyase (2SL) that replaces yxeK in some yxe gene clusters. 2SL genes from Enterococcus italicus and Dickeya dadantii complement B. subtilis yxeK mutants. We also determined that recombinant 2SL enzymes efficiently break down 2SNAC into fumarate and N-acetylcysteine, can perform the reverse reaction, and have minor activity against 2SC and other small molecule thiols. The strong preferences both YxeK and 2SL enzymes have for 2SNAC indicate that 2SC acetylation is a conserved breakdown step. The identification of a second naturally occurring 2SC breakdown pathway underscores the importance of 2SC catabolism and defines a general strategy for 2SC breakdown involving acetylation, breakdown, and deacetylation.
Collapse
|