1
|
He J, Hewett SJ. Nrf2 Regulates Basal Glutathione Production in Astrocytes. Int J Mol Sci 2025; 26:687. [PMID: 39859401 PMCID: PMC11765531 DOI: 10.3390/ijms26020687] [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: 11/11/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Astrocytes produce and export glutathione (GSH), an important thiol antioxidant essential for protecting neural cells from oxidative stress and maintaining optimal brain health. While it has been established that oxidative stress increases GSH production in astrocytes, with Nrf2 acting as a critical transcription factor regulating key components of the GSH synthetic pathway, the role of Nrf2 in controlling constitutive GSH synthetic and release mechanisms remains incompletely investigated. Our data show that naïve primary mouse astrocytes cultured from the cerebral cortices of Nrf2 knockout (Nrf2-/-) pups have significantly less intracellular and extracellular GSH levels when compared to astrocytes cultured from Nrf2 wild-type (Nrf2+/+) pups. Key components of the GSH synthetic pathway, including xCT (the substrate-specific light chain of the substrate-importing transporter, system xc-), glutamate-cysteine ligase [catalytic (GCLc) and modifying (GCLm) subunits], were affected. To wit: qRT-PCR analysis demonstrates that naïve Nrf2-/- astrocytes have significantly lower basal mRNA levels of xCT and both GCL subunits compared to naïve Nrf2+/+ astrocytes. No change in mRNA levels of glutathione synthetase (GS) or the GSH exporting transporter, Mrp1, was found. Western blot analysis reveals reduced protein levels of both subunits of GCL, while (seleno)cystine uptake into Nrf2-/- astrocytes was reduced compared to Nrf2+/+ astrocytes, confirming decreased system xc- activity. These findings suggest that Nrf2 regulates the basal production of GSH in astrocytes through constitutive transcriptional regulation of GCL and xCT.
Collapse
Affiliation(s)
| | - Sandra J. Hewett
- Program in Neuroscience, Department of Biology, Syracuse University, Syracuse, NY 13210, USA;
| |
Collapse
|
2
|
Ulusu NN. Revealing the secrets of Blue Zones. Front Pharmacol 2024; 15:1428111. [PMID: 39726786 PMCID: PMC11669513 DOI: 10.3389/fphar.2024.1428111] [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: 05/05/2024] [Accepted: 11/14/2024] [Indexed: 12/28/2024] Open
Abstract
Aging is influenced by cellular senescence mechanisms that are associated with oxidative stress. Oxidative stress is the imbalance between antioxidants and free radicals. This imbalance affects enzyme activities and causes mitochondrial dysfunction. It also slows down cellular energy production and disrupts cellular homeostasis. Additionally, oxidative stress stimulates inflammation, increases the number of point mutations, and alters intercellular communication. It can lead to epigenetic alterations, genomic instability, telomere attrition, and loss of proteostasis. Ultimately, these factors contribute to aging and the development of chronic diseases. Glucose-6-phosphate dehydrogenase (G6PD) is an antioxidant enzyme that protects cells from oxidative and nitrosative damage. It helps restore redox balance, preserve macromolecule function, and rescue cells from cellular senescence, autophagy, and stress-induced apoptosis. G6PD is considered an anti-senescence enzyme. The World Health Organization classifies G6PD variants into five groups based on the enzyme's residual activity. The first four classes are categorized according to the degree of G6PD deficiency, while the fifth class includes variants with enzyme activities greater than normal. Increased G6PD activity does not exhibit clinical manifestations. Consequently, the full spectrum of mutations and the prevalence of increased G6PD activity in the population remain unknown. The world's oldest and healthiest people live in Blue Zones. These comprise isolated populations, and there may be a geographic prevalence of high-activity G6PD variants that protect against oxidative stress-induced senescence. To uncover the secret of centenarians' longevity, additional research is needed to determine whether the hidden factor is the increased activity of the G6PD enzyme.
Collapse
Affiliation(s)
- N. Nuray Ulusu
- Department of Medical Biochemistry, School of Medicine, Koc University, Istanbul, Türkiye
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| |
Collapse
|
3
|
Park SY, Kim KY, Gwak DS, Shin SY, Jun DY, Kim YH. L-Cysteine mitigates ROS-induced apoptosis and neurocognitive deficits by protecting against endoplasmic reticulum stress and mitochondrial dysfunction in mouse neuronal cells. Biomed Pharmacother 2024; 180:117538. [PMID: 39393330 DOI: 10.1016/j.biopha.2024.117538] [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: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/13/2024] Open
Abstract
Oxidative stress and mitochondrial dysfunction play critical roles in neurodegenerative diseases. Glutathione (GSH), a key brain antioxidant, helps to neutralize reactive oxygen species (ROS) and maintain redox balance. We investigated the effectiveness of L-cysteine (L-Cys) in preventing apoptosis induced by the ROS generator 2,3-dimethoxy-1,4-naphthoquinone (DMNQ) in mouse hippocampal neuronal HT22 cells, as well as alleviating memory and cognitive impairments caused by the GSH synthesis inhibitor L-buthionine sulfoximine (BSO) in mice. DMNQ-induced apoptotic events in HT22 cells, including elevated cytosolic and mitochondrial ROS levels, DNA fragmentation, endoplasmic reticulum stress, and mitochondrial damage-mediated apoptotic pathways were dose-dependently abrogated by L-Cys (0.5-2 mM). The reduced intracellular GSH level, caused by DMNQ treatment, was restored by L-Cys cotreatment. Although L-Cys did not significantly restore GSH in the presence of BSO, it prevented DMNQ-induced ROS elevation, mitochondrial damage, and apoptosis. Furthermore, compared to N-acetylcysteine and GSH, L-Cys had higher 2,2-diphenyl-1-picrylhydrazyl and 2,2-azino-bis-3-ethylbenzothiazoline-6-sulphonic acid radical-scavenging activity. L-Cys also restored mitochondrial respiration capacity in DMNQ-treated HT22 cells by reversing mitochondrial fission-fusion dynamic balance. BSO administration (500 mg/kg/day) in mice led to neuronal deficits, including memory and cognitive impairments, which were effectively mitigated by oral L-Cys (15 or 30 mg/kg/day). L-Cys also reduced BSO-induced ROS levels in the mice hippocampus and cortex. These findings suggest that even though it does not contribute to intracellular GSH synthesis, exogenous L-Cys protects neuronal cells against oxidative stress-induced mitochondrial damage and apoptosis, by acting as a ROS scavenger, which is beneficial in ameliorating neurocognitive deficits caused by oxidative stress.
Collapse
Affiliation(s)
- Shin Young Park
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Ki Yun Kim
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Dong Seol Gwak
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Soon Young Shin
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Do Youn Jun
- AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Young Ho Kim
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea.
| |
Collapse
|
4
|
Majumder A, Bano S, Nayak KB. The Pivotal Role of One-Carbon Metabolism in Neoplastic Progression During the Aging Process. Biomolecules 2024; 14:1387. [PMID: 39595564 PMCID: PMC11591851 DOI: 10.3390/biom14111387] [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: 09/18/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
One-carbon (1C) metabolism is a complex network of metabolic reactions closely related to producing 1C units (as methyl groups) and utilizing them for different anabolic processes, including nucleotide synthesis, methylation, protein synthesis, and reductive metabolism. These pathways support the high proliferative rate of cancer cells. While drugs that target 1C metabolism (like methotrexate) have been used for cancer treatment, they often have significant side effects. Therefore, developing new drugs with minimal side effects is necessary for effective cancer treatment. Methionine, glycine, and serine are the main three precursors of 1C metabolism. One-carbon metabolism is vital not only for proliferative cells but also for non-proliferative cells in regulating energy homeostasis and the aging process. Understanding the potential role of 1C metabolism in aging is crucial for advancing our knowledge of neoplastic progression. This review provides a comprehensive understanding of the molecular complexities of 1C metabolism in the context of cancer and aging, paving the way for researchers to explore new avenues for developing advanced therapeutic interventions for cancer.
Collapse
Affiliation(s)
- Avisek Majumder
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| | - Shabana Bano
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| | - Kasturi Bala Nayak
- Quantitative Biosciences Institute, Department of Medicine, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
5
|
Al-Hatou M, Safan AS, Atta MA, Siddiqi M. Clinical and Biochemical Analysis of Glutamate-Cysteine Ligase Deficiency Presented with Late-Onset Spinocerebellar Ataxia and Hemolytic Anemia. Mol Syndromol 2024; 15:432-436. [PMID: 39359943 PMCID: PMC11444706 DOI: 10.1159/000538225] [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: 11/23/2023] [Accepted: 03/05/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Glutamate-cysteine ligase catalytic subunit (GCLC), previously known as gamma-glutamyl-cysteine synthetase, is an essential rate-limiting step in glutathione synthesis. Glutathione modulates multitudes of critical cellular processes and scavenges free radicals. Its deficiency is reported to cause hemolysis of variable severity and is a rare cause of neurological abnormalities such as spinocerebellar ataxia. Clinical Presentation We report a 55-year-old female patient with progressive late-onset ataxia, lower limb spasticity, and chronic hemolytic anemia found to have a GCLC pathogenic variant and low glutathione level. Magnetic resonance imaging of the head and cervical spine showed global cerebellar atrophy with widened folia and decreased diameter of the upper cervical spine. Blood workup revealed hemolytic anemia with genetic testing confirmed a homozygous variant, c.514 T>A in exon 4 of the GCLC gene, resulting in Ser172Thr (TCC>ACC). Management encompassed a multidisciplinary approach with a trial of high-dose alpha-lipoic acid, glutathione supplement, and physical therapy. Conclusions GCLC deficiency manifesting with hemolysis has been reported in 12 cases worldwide from 6 independent families, with only 4 cases having additional neurological manifestations. To date, no specific GCLC gene mutation has been attributed to the reported neurological constellation of symptoms. To the best of our knowledge, this is the first case report of late-onset spinocerebellar degeneration as a manifestation of c.514T>A (p. S172T) GCLC pathological variant genetic mutation.
Collapse
Affiliation(s)
- Mohammed Al-Hatou
- Department of Neurology, Neurosciences Institute, Hamad Medical Corporation, Doha, Qatar
| | - Abeer Sabry Safan
- Department of Neurology, Neurosciences Institute, Hamad Medical Corporation, Doha, Qatar
| | - Mohamed A. Atta
- Department of Internal Medicine, Hamad Medical Corporation, Doha, Qatar
| | - Maria Siddiqi
- Department of Neurology, Neurosciences Institute, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
6
|
Reyes-Soto CY, Ramírez-Carreto RJ, Ortíz-Alegría LB, Silva-Palacios A, Zazueta C, Galván-Arzate S, Karasu Ç, Túnez I, Tinkov AA, Aschner M, López-Goerne T, Chavarría A, Santamaría A. S-allyl-cysteine triggers cytotoxic events in rat glioblastoma RG2 and C6 cells and improves the effect of temozolomide through the regulation of oxidative responses. Discov Oncol 2024; 15:272. [PMID: 38977545 PMCID: PMC11231126 DOI: 10.1007/s12672-024-01145-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive form of cancer affecting the Central Nervous System (CNS) of thousands of people every year. Redox alterations have been shown to play a key role in the development and progression of these tumors as Reactive Oxygen Species (ROS) formation is involved in the modulation of several signaling pathways, transcription factors, and cytokine formation. The second-generation oral alkylating agent temozolomide (TMZ) is the first-line chemotherapeutic drug used to treat of GBM, though patients often develop primary and secondary resistance, reducing its efficacy. Antioxidants represent promising and potential coadjutant agents as they can reduce excessive ROS formation derived from chemo- and radiotherapy, while decreasing pharmacological resistance. S-allyl-cysteine (SAC) has been shown to inhibit the proliferation of several types of cancer cells, though its precise antiproliferative mechanisms remain poorly investigated. To date, SAC effects have been poorly explored in GBM cells. Here, we investigated the effects of SAC in vitro, either alone or in combination with TMZ, on several toxic and modulatory endpoints-including oxidative stress markers and transcriptional regulation-in two glioblastoma cell lines from rats, RG2 and C6, to elucidate some of the biochemical and cellular mechanisms underlying its antiproliferative properties. SAC (1-750 µM) decreased cell viability in both cell lines in a concentration-dependent manner, although C6 cells were more resistant to SAC at several of the tested concentrations. TMZ also produced a concentration-dependent effect, decreasing cell viability of both cell lines. In combination, SAC (1 µM or 100 µM) and TMZ (500 µM) enhanced the effects of each other. SAC also augmented the lipoperoxidative effect of TMZ and reduced cell antioxidant resistance in both cell lines by decreasing the TMZ-induced increase in the GSH/GSSG ratio. In RG2 and C6 cells, SAC per se had no effect on Nrf2/ARE binding activity, while in RG2 cells TMZ and the combination of SAC + TMZ decreased this activity. Our results demonstrate that SAC, alone or in combination with TMZ, exerts antitumor effects mediated by regulatory mechanisms of redox activity responses. SAC is also a safe drug for testing in other models as it produces non-toxic effects in primary astrocytes. Combined, these effects suggest that SAC affords antioxidant properties and potential antitumor efficacy against GBM.
Collapse
Affiliation(s)
- Carolina Y Reyes-Soto
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, 06726, Mexico City, Mexico
| | - Ricardo J Ramírez-Carreto
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, 06726, Mexico City, Mexico
- Facultad de Química, Universidad Nacional Autónoma de México, 04510, Mexico, Mexico
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Luz Belinda Ortíz-Alegría
- Laboratorio de Inmunología Experimental, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, 04530, Mexico City, Mexico
| | - Alejandro Silva-Palacios
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, SSA, 14080, Mexico City, Mexico
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, SSA, 14080, Mexico City, Mexico
| | - Sonia Galván-Arzate
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, S.S, 14269, Mexico City, Mexico
| | - Çimen Karasu
- Department of Medical Pharmacology, Cellular Stress Response and Signal Transduction Research Laboratory, Faculty of Medicine, Gazi University, 06500, Ankara, Turkey
| | - Isaac Túnez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Instituto de Investigaciones Biomédicas Maimónides de Córdoba (IMIBIC)Universidad de CórdobaRed Española de Excelencia en Estimulación Cerebral (REDESTIM), 14071, Córdoba, Spain
| | - Alexey A Tinkov
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
- Departament of Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
- Laboratory of Molecular Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Tessy López-Goerne
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, 04960, Mexico City, Mexico
| | - Anahí Chavarría
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, 06726, Mexico City, Mexico.
| | - Abel Santamaría
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, 04960, Mexico City, Mexico.
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
7
|
Liang LP, Sri Hari A, Day BJ, Patel M. Pharmacological elevation of glutathione inhibits status epilepticus-induced neuroinflammation and oxidative injury. Redox Biol 2024; 73:103168. [PMID: 38714094 PMCID: PMC11087235 DOI: 10.1016/j.redox.2024.103168] [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: 03/20/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/09/2024] Open
Abstract
Glutathione (GSH) is a major endogenous antioxidant, and its depletion has been observed in several brain diseases including epilepsy. Previous studies in our laboratory have shown that dimercaprol (DMP) can elevate GSH via post-translational activation of glutamate cysteine ligase (GCL), the rate limiting GSH biosynthetic enzyme and inhibit neuroinflammation in vitro. Here we determined 1) the role of cysteamine as a new mechanism by which DMP increases GSH biosynthesis and 2) its ability to inhibit neuroinflammation and neuronal injury in the rat kainate model of epilepsy. DMP depleted cysteamine in a time- and concentration-dependent manner in a cell free system. To guide the in vivo administration of DMP, its pharmacokinetic profile was determined in the plasma, liver, and brain. The results confirmed DMP's ability to cross the blood-brain-barrier. Treatment of rats with DMP (30 mg/kg) depleted cysteamine in the liver and hippocampus that was associated with increased GCL activity in these tissues. GSH levels were significantly increased (20 %) in the hippocampus 1 h after 30 mg/kg DMP administration. Following DMP (30 mg/kg) administration once daily, a marked attenuation of GSH depletion was seen in the SE model. SE-induced inflammatory markers including cytokine release, microglial activation, and neuronal death were significantly attenuated in the hippocampus with DMP treatment. Taken together, these results highlight the importance of restoring redox status with rescue of GSH depletion by DMP in post epileptogenic insults.
Collapse
Affiliation(s)
- Li-Ping Liang
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ashwini Sri Hari
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Brian J Day
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA; Department of Medicine, National Jewish Health, Denver, CO, 80202, USA
| | - Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
8
|
Fogarty EF, Harch PG. Case report: Dementia sensitivity to altitude changes and effective treatment with hyperbaric air and glutathione precursors. Front Neurol 2024; 15:1356662. [PMID: 38978816 PMCID: PMC11229546 DOI: 10.3389/fneur.2024.1356662] [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: 12/15/2023] [Accepted: 05/10/2024] [Indexed: 07/10/2024] Open
Abstract
A 78-year-old man with dementia experienced waxing and waning of symptoms with changes in altitude as he traveled from his home in the Rocky Mountains to lower elevations and back. To replicate the improvement in his symptoms with travel to lower elevations (higher pressure), the patient was treated with a near-identical repressurization in a hyperbaric chamber using compressed air. With four 1-h treatments at 1.3 Atmospheres Absolute (ATA) and concurrent administration of low-dose oral glutathione amino acid precursors, he recovered speech and showed improvement in activities of daily living. Regional broadcast media had documented his novel recovery. Nosocomial COVID-19 and withdrawal of hyperbaric air therapy led to patient demise 7 months after initiation of treatment. It is theorized that hyperbaric air therapy stimulated mitochondrial biochemical and physical changes, which led to clinical improvement.
Collapse
Affiliation(s)
| | - Paul G. Harch
- LSU Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
9
|
Hu K, Xu Y, Fan J, Liu H, Di C, Xu F, Wu L, Ding K, Zhang T, Wang L, Ai H, Xie L, Wang G, Liang Y. Feasibility exploration of GSH in the treatment of acute hepatic encephalopathy from the aspects of pharmacokinetics, pharmacodynamics, and mechanism. Front Pharmacol 2024; 15:1387409. [PMID: 38887546 PMCID: PMC11181355 DOI: 10.3389/fphar.2024.1387409] [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: 02/17/2024] [Accepted: 05/06/2024] [Indexed: 06/20/2024] Open
Abstract
Our previous study highlighted the therapeutic potential of glutathione (GSH), an intracellular thiol tripeptide ubiquitous in mammalian tissues, in mitigating hepatic and cerebral damage. Building on this premise, we posited the hypothesis that GSH could be a promising candidate for treating acute hepatic encephalopathy (AHE). To verify this conjecture, we systematically investigated the feasibility of GSH as a therapeutic agent for AHE through comprehensive pharmacokinetic, pharmacodynamic, and mechanistic studies using a thioacetamide-induced AHE rat model. Our pharmacodynamic data demonstrated that oral GSH could significantly improve behavioral scores and reduce hepatic damage of AHE rats by regulating intrahepatic ALT, AST, inflammatory factors, and homeostasis of amino acids. Additionally, oral GSH demonstrated neuroprotective effects by alleviating the accumulation of intracerebral glutamine, down-regulating glutamine synthetase, and reducing taurine exposure. Pharmacokinetic studies suggested that AHE modeling led to significant decrease in hepatic and cerebral exposure of GSH and cysteine. However, oral GSH greatly enhanced the intrahepatic and intracortical GSH and CYS in AHE rats. Given the pivotal roles of CYS and GSH in maintaining redox homeostasis, we investigated the interplay between oxidative stress and pathogenesis/treatment of AHE. Our data revealed that GSH administration significantly relieved oxidative stress levels caused by AHE modeling via down-regulating the expression of NADPH oxidase 4 (NOX4) and NF-κB P65. Importantly, our findings further suggested that GSH administration significantly regulated the excessive endoplasmic reticulum (ER) stress caused by AHE modeling through the iNOS/ATF4/Ddit3 pathway. In summary, our study uncovered that exogenous GSH could stabilize intracerebral GSH and CYS levels to act on brain oxidative and ER stress, which have great significance for revealing the therapeutic effect of GSH on AHE and promoting its further development and clinical application.
Collapse
Affiliation(s)
- Kangrui Hu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yexin Xu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jiye Fan
- Department of Pharmacy, Hebei Chemical and Pharmaceutical College, Shijiazhuang, Hebei Province, China
| | - Huafang Liu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Chanjuan Di
- Hebei Zhitong Biopharmaceutical Co., Ltd., Gucheng, Hebei Province, China
| | - Feng Xu
- Hebei Zhitong Biopharmaceutical Co., Ltd., Gucheng, Hebei Province, China
| | - Linlin Wu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ke Ding
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tingting Zhang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Leyi Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haoyu Ai
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Lin Xie
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | | | - Yan Liang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
10
|
Liddell JR, Hilton JBW, Kysenius K, Billings JL, Nikseresht S, McInnes LE, Hare DJ, Paul B, Mercer SW, Belaidi AA, Ayton S, Roberts BR, Beckman JS, McLean CA, White AR, Donnelly PS, Bush AI, Crouch PJ. Microglial ferroptotic stress causes non-cell autonomous neuronal death. Mol Neurodegener 2024; 19:14. [PMID: 38317225 PMCID: PMC10840184 DOI: 10.1186/s13024-023-00691-8] [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: 10/20/2023] [Accepted: 11/28/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Ferroptosis is a form of regulated cell death characterised by lipid peroxidation as the terminal endpoint and a requirement for iron. Although it protects against cancer and infection, ferroptosis is also implicated in causing neuronal death in degenerative diseases of the central nervous system (CNS). The precise role for ferroptosis in causing neuronal death is yet to be fully resolved. METHODS To elucidate the role of ferroptosis in neuronal death we utilised co-culture and conditioned medium transfer experiments involving microglia, astrocytes and neurones. We ratified clinical significance of our cell culture findings via assessment of human CNS tissue from cases of the fatal, paralysing neurodegenerative condition of amyotrophic lateral sclerosis (ALS). We utilised the SOD1G37R mouse model of ALS and a CNS-permeant ferroptosis inhibitor to verify pharmacological significance in vivo. RESULTS We found that sublethal ferroptotic stress selectively affecting microglia triggers an inflammatory cascade that results in non-cell autonomous neuronal death. Central to this cascade is the conversion of astrocytes to a neurotoxic state. We show that spinal cord tissue from human cases of ALS exhibits a signature of ferroptosis that encompasses atomic, molecular and biochemical features. Further, we show the molecular correlation between ferroptosis and neurotoxic astrocytes evident in human ALS-affected spinal cord is recapitulated in the SOD1G37R mouse model where treatment with a CNS-permeant ferroptosis inhibitor, CuII(atsm), ameliorated these markers and was neuroprotective. CONCLUSIONS By showing that microglia responding to sublethal ferroptotic stress culminates in non-cell autonomous neuronal death, our results implicate microglial ferroptotic stress as a rectifiable cause of neuronal death in neurodegenerative disease. As ferroptosis is currently primarily regarded as an intrinsic cell death phenomenon, these results introduce an entirely new pathophysiological role for ferroptosis in disease.
Collapse
Affiliation(s)
- Jeffrey R Liddell
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - James B W Hilton
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kai Kysenius
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jessica L Billings
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Sara Nikseresht
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Lachlan E McInnes
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Dominic J Hare
- Atomic Medicine Initiative, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Bence Paul
- School of Earth Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Stephen W Mercer
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Abdel A Belaidi
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Scott Ayton
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Blaine R Roberts
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Joseph S Beckman
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | - Catriona A McLean
- Anatomical Pathology, Alfred Hospital, Melbourne, VIC, 3005, Australia
| | - Anthony R White
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ashley I Bush
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Peter J Crouch
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
11
|
Tripathi AS, Fatima N, Tripathi P, Tripathi R, Alka, Zaki MEA, Mohapatra L, Yasir M, Maurya RK. Beneficial effect of 5-HT1b/1d agonist on Parkinson's disease by modulating glutamate and reducing deposition of α-synuclein. J Biochem Mol Toxicol 2024; 38:e23627. [PMID: 38229316 DOI: 10.1002/jbt.23627] [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: 07/21/2023] [Revised: 11/13/2023] [Accepted: 12/14/2023] [Indexed: 01/18/2024]
Abstract
The given investigation examined the neuroprotection role of 5-HT1b/1d agonist in reserpine induced Parkinson's disease (PD) in male Wistar rats. PD was induced in rats by reserpine at 5 mg/kg ip for 3 days and thereafter the rats were provided with the following treatments for 4 days, zolmitriptan (ZLM) group (30 mg/kg ip); STD group (levodopa + carbidopa, 200 + 5 mg/kg ip); ZLM + GA group (zolmitriptan, 30 mg/kg ip and glutamic acid, 1.5 mg/kg); ZLM + DX group (zolmitriptan, 30 mg/kg ip and dextromethorphan, 20 mg/kg ip). All the groups were then assessed for cognitive and motor functions at the end of the protocol. Moreover, oxidative stress parameters and histopathological changes were observed in rats of all treatment groups. Deposition of α-synuclein in the brain tissue was observed by silver staining. Data of this investigation revealed that motor and cognitive functions were improved in the ZLM-treated group compared with the negative control group, which was observed to be reversed in ZLM + GA group. Treatment with ZLM ameliorated oxidative stress and histopathological changes in the brain tissue of PD rats. Further, ZLM reduced the deposition of α-synuclein in PD rats, which reversed in ZLM + GA-treated group. This study concludes by stating that 5-HT1b/1d agonist can prevent neurodegeneration and reduce oxidative stress in PD rats. The probable underlying mechanism of such an effect of 5-HT1b/1d agonist could be by regulating the deposition of α-synuclein and reducing the expression of NMDA receptor.
Collapse
Affiliation(s)
- Alok Shiomurti Tripathi
- Department of Pharmacology, Era College of Pharmacy, ERA University, Lucknow, Uttar Pradesh, India
| | - Needa Fatima
- Department of Pharmacology, Amity Institute of Pharmacy, Lucknow, Amity University, Noida, Uttar Pradesh, India
| | - Pankaj Tripathi
- Department of Pharmacology, Nootan Pharmacy College, Sankalchand Patel University, Visnagar, Gujarat, India
| | - Rina Tripathi
- Department of Pharmacology, Nootan Pharmacy College, Sankalchand Patel University, Visnagar, Gujarat, India
| | - Alka
- Department of Pharmacology, Amity Institute of Pharmacy, Lucknow, Amity University, Noida, Uttar Pradesh, India
| | - Magdi E A Zaki
- Department of Chemistry, College of Science, Imam Mohammad lbn Saud Islamic University, Riyadh, Saudi Arabia
| | - Lucy Mohapatra
- Department of Pharmacology, Amity Institute of Pharmacy, Lucknow, Amity University, Noida, Uttar Pradesh, India
| | - Mohammad Yasir
- Department of Pharmacology, Amity Institute of Pharmacy, Lucknow, Amity University, Noida, Uttar Pradesh, India
| | - Rahul K Maurya
- Department of Pharmacology, Amity Institute of Pharmacy, Lucknow, Amity University, Noida, Uttar Pradesh, India
| |
Collapse
|
12
|
Cui D, Chen Y, Ye B, Guo W, Wang D, He J. Natural products for the treatment of neurodegenerative diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155101. [PMID: 37778246 DOI: 10.1016/j.phymed.2023.155101] [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: 01/29/2023] [Revised: 08/29/2023] [Accepted: 09/17/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Neurodegenerative diseases are among the most common diseases in older adults worldwide. Alzheimer's disease (AD) and Parkinson's disease (PD) are two of the most common neurodegenerative diseases, and are accompanied by cerebral cortical atrophy, neuronal loss, protein accumulation, and excessive accumulation of metal ions. Natural products exhibit outstanding performance in improving cerebral circulatory disorders, promoting cerebral haematoma absorption, repairing damaged nerve tissue, and improving damaged nerve function. In recent years, studies have shown that neuroinflammatory mechanisms and signalling pathways closely related to the occurrence and development of neurological diseases include microglial activation, nuclear factor-κB (NF-κB) pathway, mitogen activated protein kinases (MAPK) pathway, reactive oxygen pathway, nucleotide binding oligomerisation domain-like receptor protein3 (NLRP3) inflammasomes, toll-like receptor4 (TLR4) pathway, nuclear factor erythroid 2-related factor 2 (Nrf2)/hemeoxygenase-1 (HO-1) pathway, phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, and intestinal flora. Therefore, this study considered the mechanism of neurological diseases as the starting point to review the mechanism of action of natural products in the prevention and treatment of AD and PD in recent years to provide a theoretical basis for clinical prevention and treatment. AIM Natural products are a promising source of novel lead structures that have long been used to treat various nervous system diseases. METHODOLOGY This review collected literature on neurological diseases and natural products from 2012 to 2022, which were mainly searched through databases such as ScienceDirect, Springer, PubMed, SciFinder, China National Knowledge Infrastructure (CNKI), Wanfang, Google Scholar, and Baidu Academic. The following keywords were searched: neurological disorders, natural products, signalling pathway, mechanism of action. RESULTS This review summarises the pathogenesis of degenerative neurological diseases, recent findings on natural products used in neurodegenerative diseases, and the molecular mechanisms underlying these effects.
Collapse
Affiliation(s)
- Donghan Cui
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University, West China Hospital, Chengdu 610041, China
| | - Yajuan Chen
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Bengui Ye
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China; Medical College of Tibet University, Lasa 850002, China
| | - Wenhao Guo
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University, West China Hospital, Chengdu 610041, China.
| | - Dongdong Wang
- Centre for Metabolism, Obesity, and Diabetes Research, Department of Medicine, McMaster University, HSC 4N71, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada.
| | - Jun He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China.
| |
Collapse
|
13
|
Adekomi DA, Olajide OJ, Adewale OO, Okesina AA, Fatoki JO, Falana BA, Adeniyi TD, Adegoke AA, Ojo WA, Alabi SO. D-ribose-L-cysteine exhibits neuroprotective activity through inhibition of oxido-behavioral dysfunctions and modulated activities of neurotransmitters in the cerebellum of Juvenile mice exposed to ethanol. Drug Chem Toxicol 2023; 46:746-756. [PMID: 35723231 DOI: 10.1080/01480545.2022.2088783] [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: 07/23/2021] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/03/2022]
Abstract
Alcohol exposure to the cerebellum has been known to trigger cerebellar dysfunctions through several mechanisms. This present study was designed to evaluate the repealing effect of D-ribose-L-cysteine (DRLC) on alcohol-induced cerebellar dysfunctions in juvenile BALB/c mice. The animals were randomly divided into 4 groups (n = 10 per group). Mice were given oral administration of normal saline (control), DRLC (100 mg/kg, p.o), ethanol (0.2 mL of 10% w/v), or DRLC (100 mg/kg, p.o) + ethanol (0.2 mL of 10% w/v). On day 29 of the study (i.e., 24 h after the administration of the last respective doses), neurochemical quantification of the respective levels of serotonin and dopamine, lipid peroxidation, total antioxidant, superoxide dismutase, and glutathione peroxidase in the cerebellar tissues of the mice were analyzed. Compared with the saline-treated group, the studied neurochemical indices were modulated across the various experimental groups. The administration of ethanol significantly modulates the levels of monoamine neurotransmitters (serotonin and dopamine) as well as contents of total antioxidants, activities of superoxide dismutase, and glutathione peroxidase, with a concurrently increased level of lipid peroxidase in the cerebellar tissue of the mice. DRLC significantly reverses these effects in the DRLC + ethanol co-treated group. Combined exposure to DRLC + ethanol counteracts the deleterious effect of ethanol in the cerebellum of juvenile BALB/c mice via monoamine neurotransmitter, lipid peroxidation, total antioxidant status, superoxide dismutase, and glutathione peroxidase action pathways. Therefore, DRLC could be a pharmacologic or therapeutic agent in attenuating the deleterious effects of alcohol on the cerebellum.
Collapse
Affiliation(s)
- Damilare Adedayo Adekomi
- Department of Anatomy, Neuroscience and Cell Biology Unit, Osun State University, Osogbo, Nigeria
| | - Olamide Janet Olajide
- Department of Anatomy, Neuroscience and Cell Biology Unit, Osun State University, Osogbo, Nigeria
| | - Omowumi Oyeronke Adewale
- Department of Biochemistry, Faculty of Basic and Applied Sciences, Osun State University, Osogbo, Nigeria
| | | | - John Olabode Fatoki
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, Osun State University, Osogbo, Nigeria
| | - Benedict Abiola Falana
- Department of Anatomy, Neuroscience and Cell Biology Unit, Osun State University, Osogbo, Nigeria
| | - Temidayo Daniel Adeniyi
- Department of Medical Laboratory Science, Faculty of Allied Health Science, University of Medical Sciences, Ondo State, Nigeria
| | | | - Waliu Adetunji Ojo
- Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomosho, Nigeria
| | | |
Collapse
|
14
|
Huang Y, Li X, Zhang Z, Xiong L, Wang Y, Wen Y. Photodynamic Therapy Combined with Ferroptosis Is a Synergistic Antitumor Therapy Strategy. Cancers (Basel) 2023; 15:5043. [PMID: 37894410 PMCID: PMC10604985 DOI: 10.3390/cancers15205043] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/20/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Ferroptosis is a programmed death mode that regulates redox homeostasis in cells, and recent studies suggest that it is a promising mode of tumor cell death. Ferroptosis is regulated by iron metabolism, lipid metabolism, and intracellular reducing substances, which is the mechanism basis of its combination with photodynamic therapy (PDT). PDT generates reactive oxygen species (ROS) and 1O2 through type I and type II photochemical reactions, and subsequently induces ferroptosis through the Fenton reaction and the peroxidation of cell membrane lipids. PDT kills tumor cells by generating excessive cytotoxic ROS. Due to the limited laser depth and photosensitizer enrichment, the systemic treatment effect of PDT is not good. Combining PDT with ferroptosis can compensate for these shortcomings. Nanoparticles constructed by photosensitizers and ferroptosis agonists are widely used in the field of combination therapy, and their targeting and biological safety can be improved through modification. These nanoparticles not only directly kill tumor cells but also further exert the synergistic effect of PDT and ferroptosis by activating antitumor immunity, improving the hypoxia microenvironment, and inhibiting the tumor angiogenesis. Ferroptosis-agonist-induced chemotherapy and PDT-induced ablation also have good clinical application prospects. In this review, we summarize the current research progress on PDT and ferroptosis and how PDT and ferroptosis promote each other.
Collapse
Affiliation(s)
- Yunpeng Huang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Xiaoyu Li
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China;
| | - Zijian Zhang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Li Xiong
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Yongxiang Wang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Yu Wen
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| |
Collapse
|
15
|
Xu X, Wang SS, Zhang L, Lu AX, Lin Y, Liu JX, Yan CH. Methylmercury induced ferroptosis by interference of iron homeostasis and glutathione metabolism in CTX cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 335:122278. [PMID: 37517642 DOI: 10.1016/j.envpol.2023.122278] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 08/01/2023]
Abstract
Environmental methylmercury (MeHg) exposure has gained global attention owing to its serious health hazards, especially neurotoxicity. Ferroptosis is a novel form of programmed cell death characterized by lipid peroxidation and iron overload. However, the occurrence of ferroptosis and its underlying mechanisms have not been fully elucidated in the methylmercury-induced neurotoxicity and the role of Nrf2 in MeHg-induced ferroptosis remains unexplored. In this study, we verified that MeHg decreased cell viability in a dose- and time-dependent manner in the Rat Brain Astrocytes cells (CTX cells). MeHg (3.5 μmol/L) exposure induced CTX cells to undergo ferroptosis, as evidenced by glutathione (GSH) depletion, lipid peroxidation, and iron overload, which was significantly rescued by the ferroptosis-specific inhibitors Ferrostatin-1 and Deferoxamine. MeHg directly disrupted the process of GSH metabolism by downregulating of SLC7A11 and GPX4 and interfered with intracellular iron homeostasis through inhibition of iron storage and export. Simultaneously, the expression of Nrf2 was upregulated by MeHg in CTX cells. Hence, the inhibition of Nrf2 activity further downregulated the levels of GPX4, SLC7A11, FTH1, and SLC40A1, which aggravated MeHg-induced ferroptosis to a greater extent. Overall, our findings provided evidence that ferroptosis played a critical role in MeHg-induced neurotoxicity, and suppressing Nrf2 activity further exacerbated MeHg-induced ferroptosis in CTX cells.
Collapse
Affiliation(s)
- Xi Xu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Su-Su Wang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - An-Xin Lu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yin Lin
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun-Xia Liu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chong-Huai Yan
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
16
|
Kamrani-Sharif R, Hayes AW, Gholami M, Salehirad M, Allahverdikhani M, Motaghinejad M, Emanuele E. Oxytocin as neuro-hormone and neuro-regulator exert neuroprotective properties: A mechanistic graphical review. Neuropeptides 2023; 101:102352. [PMID: 37354708 DOI: 10.1016/j.npep.2023.102352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 03/28/2023] [Accepted: 06/12/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Neurodegeneration is progressive cell loss in specific neuronal populations, often resulting in clinical consequences with significant medical, societal, and economic implications. Because of its antioxidant, anti-inflammatory, and anti-apoptotic properties, oxytocin has been proposed as a potential neuroprotective and neurobehavioral therapeutic agent, including modulating mood disturbances and cognitive enchantment. METHODS Literature searches were conducted using the following databases Web of Science, PubMed, Elsevier Science Direct, Google Scholar, the Core Collection, and Cochrane from January 2000 to February 2023 for articles dealing with oxytocin neuroprotective properties in preventing or treating neurodegenerative disorders and diseases with a focus on oxidative stress, inflammation, and apoptosis/cell death. RESULTS The neuroprotective effects of oxytocin appears to be mediated by its anti-inflammatory properties, inhibition of neuro inflammation, activation of several antioxidant enzymes, inhibition of oxidative stress and free radical formation, activation of free radical scavengers, prevent of mitochondrial dysfunction, and inhibition of apoptosis. CONCLUSION Oxytocin acts as a neuroprotective agent by preventing neuro-apoptosis, neuro-inflammation, and neuronal oxidative stress, and by restoring mitochondrial function.
Collapse
Affiliation(s)
- Roya Kamrani-Sharif
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - A Wallace Hayes
- University of South Florida College of Public Health, Tampa, FL, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Mina Gholami
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Salehirad
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Allahverdikhani
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
17
|
Emmert S, Quargnali G, Thallmair S, Rivera-Fuentes P. A locally activatable sensor for robust quantification of organellar glutathione. Nat Chem 2023; 15:1415-1421. [PMID: 37322101 PMCID: PMC10533397 DOI: 10.1038/s41557-023-01249-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/22/2023] [Indexed: 06/17/2023]
Abstract
Glutathione (GSH) is the main determinant of intracellular redox potential and participates in multiple cellular signalling pathways. Achieving a detailed understanding of intracellular GSH homeostasis depends on the development of tools to map GSH compartmentalization and intra-organelle fluctuations. Here we present a GSH-sensing platform for live-cell imaging, termed targetable ratiometric quantitative GSH (TRaQ-G). This chemogenetic sensor possesses a unique reactivity turn-on mechanism, ensuring that the small molecule is only sensitive to GSH in a desired location. Furthermore, TRaQ-G can be fused to a fluorescent protein to give a ratiometric response. Using TRaQ-G fused to a redox-insensitive fluorescent protein, we demonstrate that the nuclear and cytosolic GSH pools are independently regulated during cell proliferation. This sensor was used in combination with a redox-sensitive fluorescent protein to quantify redox potential and GSH concentration simultaneously in the endoplasmic reticulum. Finally, by exchanging the fluorescent protein, we created a near-infrared, targetable and quantitative GSH sensor.
Collapse
Affiliation(s)
- Sarah Emmert
- Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédéral de Lausanne, Lausanne, Switzerland
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Gianluca Quargnali
- Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédéral de Lausanne, Lausanne, Switzerland
| | | | - Pablo Rivera-Fuentes
- Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédéral de Lausanne, Lausanne, Switzerland.
- Department of Chemistry, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
18
|
Zhang YM, Qi YB, Gao YN, Chen WG, Zhou T, Zang Y, Li J. Astrocyte metabolism and signaling pathways in the CNS. Front Neurosci 2023; 17:1217451. [PMID: 37732313 PMCID: PMC10507181 DOI: 10.3389/fnins.2023.1217451] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/18/2023] [Indexed: 09/22/2023] Open
Abstract
Astrocytes comprise half of the cells in the central nervous system and play a critical role in maintaining metabolic homeostasis. Metabolic dysfunction in astrocytes has been indicated as the primary cause of neurological diseases, such as depression, Alzheimer's disease, and epilepsy. Although the metabolic functionalities of astrocytes are well known, their relationship to neurological disorders is poorly understood. The ways in which astrocytes regulate the metabolism of glucose, amino acids, and lipids have all been implicated in neurological diseases. Metabolism in astrocytes has also exhibited a significant influence on neuron functionality and the brain's neuro-network. In this review, we focused on metabolic processes present in astrocytes, most notably the glucose metabolic pathway, the fatty acid metabolic pathway, and the amino-acid metabolic pathway. For glucose metabolism, we focused on the glycolysis pathway, pentose-phosphate pathway, and oxidative phosphorylation pathway. In fatty acid metabolism, we followed fatty acid oxidation, ketone body metabolism, and sphingolipid metabolism. For amino acid metabolism, we summarized neurotransmitter metabolism and the serine and kynurenine metabolic pathways. This review will provide an overview of functional changes in astrocyte metabolism and provide an overall perspective of current treatment and therapy for neurological disorders.
Collapse
Affiliation(s)
- Yong-mei Zhang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying-bei Qi
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ya-nan Gao
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wen-gang Chen
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ting Zhou
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yi Zang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jia Li
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
19
|
Juchem C, Swanberg KM, Prinsen H, Pelletier D. In vivo cortical glutathione response to oral fumarate therapy in relapsing-remitting multiple sclerosis: A single-arm open-label phase IV trial using 7-Tesla 1H MRS. Neuroimage Clin 2023; 39:103495. [PMID: 37651844 PMCID: PMC10480324 DOI: 10.1016/j.nicl.2023.103495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/29/2023] [Accepted: 08/10/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND This is an open-label, single-arm, single-center pilot study using 7-Tesla in vivo proton magnetic resonance spectroscopy (1H MRS) to measure brain cortical glutathione concentration at baseline before and during the use of oral fumarates as a disease-modifying therapy for multiple sclerosis. The primary endpoint of this research was the change in prefrontal cortex glutathione concentration relative to a therapy-naïve baseline after one year of oral fumarate therapy. METHODS Brain glutathione concentrations were examined by 1H MRS in single prefrontal and occipital cortex cubic voxels (2.5 × 2.5 × 2.5 cm3) before and during initiation of oral fumarate therapy (120 mg b.i.d. for 7 days and 240 mg b.i.d. thereafter). Additional measurements of related metabolites glutamate, glutamine, myoinositol, total N-acetyl aspartate, and total choline were also acquired in voxels centered on the same regions. Seven relapsing-remitting multiple sclerosis patients (4 f / 3 m, age range 28-50 years, mean age 40 years) naïve to fumarate therapy were scanned at pre-therapy baseline and after 1, 3, 6 and 12 months of therapy. A group of 8 healthy volunteers (4 f / 4 m, age range 33-48 years, mean age 41 years) was also scanned at baseline and Month 6 to characterize 1H-MRS measurement reproducibility over a comparable time frame. RESULTS In the multiple sclerosis cohort, general linear models demonstrated a significant positive linear relationship between prefrontal glutathione and time either linearly across all time points (+0.05 ± 0.02 mM/month, t(27) = 2.6, p = 0.02) or specifically for factor variable Month 12 (+0.6 ± 0.3 mM/12 months, t(24) = 2.2, p = 0.04) relative to baseline. No such effects of time on glutathione concentration were demonstrated in the occipital cortex or in the healthy volunteer group. Changes in occipital total choline were further observed in the multiple sclerosis cohort as well as prefrontal total choline and occipital glutamine and myoinositol in the control cohort throughout the study duration. CONCLUSIONS While the open-label single-arm pilot study design and abbreviated control series cannot support firm conclusions about the influence of oral fumarate therapy independent of test-retest factors or normal biological variation in a state of either health or disease, these results do justify further investigation at a larger scale into the potential relationship between prefrontal cortex glutathione increases and oral fumarate therapy in relapsing-remitting multiple sclerosis.
Collapse
Affiliation(s)
- Christoph Juchem
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States; Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States; Department of Biomedical Engineering, Columbia University Fu Foundation, School of Engineering and Applied Science, New York, NY, United States; Department of Radiology, Columbia University College of Physicians and Surgeons, New York, NY, United States.
| | - Kelley M Swanberg
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States; Department of Biomedical Engineering, Columbia University Fu Foundation, School of Engineering and Applied Science, New York, NY, United States
| | - Hetty Prinsen
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States
| | - Daniel Pelletier
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States; Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| |
Collapse
|
20
|
Watling SE, Rhind SG, Warsh J, Green D, McCluskey T, Tong J, Truong P, Chavez S, Richardson JD, Kish SJ, Boileau I. Exploring brain glutathione and peripheral blood markers in posttraumatic stress disorder: a combined [1H]MRS and peripheral blood study. Front Psychiatry 2023; 14:1195012. [PMID: 37333909 PMCID: PMC10272391 DOI: 10.3389/fpsyt.2023.1195012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/09/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Oxidative stress has been implicated in psychiatric disorders, including posttraumatic stress disorder (PTSD). Currently, the status of glutathione (GSH), the brain's most abundant antioxidant, in PTSD remains uncertain. Therefore, the current study investigated brain concentrations of GSH and peripheral concentrations of blood markers in individuals with PTSD vs. Healthy Controls (HC). Methods GSH spectra was acquired in the anterior cingulate cortex (ACC) and dorsolateral prefrontal cortex (DLPFC) using MEGA-PRESS, a J-difference-editing acquisition method. Peripheral blood samples were analyzed for concentrations of metalloproteinase (MMP)-9, tissue inhibitors of MMP (TIMP)-1,2, and myeloperoxidase (MPO). Results There was no difference in GSH between PTSD and HC in the ACC (n = 30 PTSD, n = 20 HC) or DLPFC (n = 14 PTSD, n = 18 HC). There were no group differences between peripheral blood markers (P > 0.3) except for (non-significantly) lower TIMP-2 in PTSD. Additionally, TIMP-2 and GSH in the ACC were positively related in those with PTSD. Finally, MPO and MMP-9 were negatively associated with duration of PTSD. Conclusions We do not report altered GSH concentrations in the ACC or DLPFC in PTSD, however, systemic MMPs and MPO might be implicated in central processes and progression of PTSD. Future research should investigate these relationships in larger sample sizes.
Collapse
Affiliation(s)
- Sarah E. Watling
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Shawn G. Rhind
- Faculty of Kinesiology & Physical Education, University of Toronto, Toronto, ON, Canada
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON, Canada
| | - Jerry Warsh
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Duncan Green
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Tina McCluskey
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Junchao Tong
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Peter Truong
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Sofia Chavez
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - J. Don Richardson
- The MacDonald Franklin Operational Stress Injury (OSI) Research Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Psychiatry, Western University, London, ON, Canada
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
- St. Joseph's London Operational Stress Injury (OSI), Parkwood Institute, St. Joseph's Health Care, London, ON, Canada
| | - Stephen J. Kish
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Isabelle Boileau
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
Sun L, Ye KX, Wong HLK, Wang L, Lim SL, Chao YX, Zhang C, Yap KZ, Feng L. The Effects of Medium Chain Triglyceride for Alzheimer's Disease Related Cognitive Impairment: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2023:JAD230406. [PMID: 37248908 PMCID: PMC10357178 DOI: 10.3233/jad-230406] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
BACKGROUND The current lack of effective drug therapies for Alzheimer's disease (AD) has prompted researchers to seek alternative nutritional therapies, such as medium chain triglycerides (MCTs). However, results are inconclusive. OBJECTIVE This systematic review and meta-analysis aims to summarize current evidence on the effect of MCT on cognitive function in patients with mild cognitive impairment (MCI) or AD. METHODS A systematic search was conducted up until December 16, 2022, to identify human interventions reporting the effects of MCT on cognitive functioning of MCI or AD patients. 995 non-duplicated publications were identified, of which nine (n = 10 studies) met the inclusion criteria. RESULTS Meta-analysis showed cognitive improvements in general (SMD = 0.64; 95% CI [0.05, 1.24]), but not in memory, language, and attention domains after oral MCT administration, compared to placebo. The effect of MCT was greater among APOEɛ4 (-) subjects than APOEɛ4 (+) subjects (SMD = 1.87; 95% CI [0.35, 3.40]). CONCLUSION This review provides some evidence that treatment with MCT could improve general cognitive function in APOEɛ4 (-) cognitive impaired patients. Better characterized clinical studies are warranted before making a definitive conclusion on the use of MCT for MCI and AD management.
Collapse
Affiliation(s)
- Lina Sun
- School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Kaisy Xinhong Ye
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Lingyan Wang
- School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Su Lin Lim
- Department of Dietetics, National University Hospital, Singapore, Singapore
| | - Yin Xia Chao
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| | - Can Zhang
- Mass General Institute for Neurodegenerative, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Kai Zhen Yap
- Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Lei Feng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
22
|
Wunsch FT, Metzler-Nolte N, Theiss C, Matschke V. Defects in Glutathione System in an Animal Model of Amyotrophic Lateral Sclerosis. Antioxidants (Basel) 2023; 12:antiox12051014. [PMID: 37237880 DOI: 10.3390/antiox12051014] [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: 03/20/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progredient neurodegenerative disease characterized by a degeneration of the first and second motor neurons. Elevated levels of reactive oxygen species (ROS) and decreased levels of glutathione, which are important defense mechanisms against ROS, have been reported in the central nervous system (CNS) of ALS patients and animal models. The aim of this study was to determine the cause of decreased glutathione levels in the CNS of the ALS model wobbler mouse. We analyzed changes in glutathione metabolism in the spinal cord, hippocampus, cerebellum, liver, and blood samples of the ALS model, wobbler mouse, using qPCR, Western Blot, HPLC, and fluorometric assays. Here, we show for the first time a decreased expression of enzymes involved in glutathione synthesis in the cervical spinal cord of wobbler mice. We provide evidence for a deficient glutathione metabolism, which is not restricted to the nervous system, but can be seen in various tissues of the wobbler mouse. This deficient system is most likely the reason for an inefficient antioxidative system and, thus, for elevated ROS levels.
Collapse
Affiliation(s)
- Franziska T Wunsch
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, D-44801 Bochum, Germany
- International Graduate School of Neuroscience (IGSN), Ruhr-University Bochum, D-44801 Bochum, Germany
| | - Nils Metzler-Nolte
- Inorganic Chemistry I-Bioinorganic Chemistry, Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, D-44801 Bochum, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, D-44801 Bochum, Germany
- International Graduate School of Neuroscience (IGSN), Ruhr-University Bochum, D-44801 Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, D-44801 Bochum, Germany
| |
Collapse
|
23
|
Karami F, Jamaati H, Coleman-Fuller N, Zeini MS, Hayes AW, Gholami M, Salehirad M, Darabi M, Motaghinejad M. Is metformin neuroprotective against diabetes mellitus-induced neurodegeneration? An updated graphical review of molecular basis. Pharmacol Rep 2023; 75:511-543. [PMID: 37093496 DOI: 10.1007/s43440-023-00469-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 04/25/2023]
Abstract
Diabetes mellitus (DM) is a metabolic disease that activates several molecular pathways involved in neurodegenerative disorders. Metformin, an anti-hyperglycemic drug used for treating DM, has the potential to exert a significant neuroprotective role against the detrimental effects of DM. This review discusses recent clinical and laboratory studies investigating the neuroprotective properties of metformin against DM-induced neurodegeneration and the roles of various molecular pathways, including mitochondrial dysfunction, oxidative stress, inflammation, apoptosis, and its related cascades. A literature search was conducted from January 2000 to December 2022 using multiple databases including Web of Science, Wiley, Springer, PubMed, Elsevier Science Direct, Google Scholar, the Core Collection, Scopus, and the Cochrane Library to collect and evaluate peer-reviewed literature regarding the neuroprotective role of metformin against DM-induced neurodegenerative events. The literature search supports the conclusion that metformin is neuroprotective against DM-induced neuronal cell degeneration in both peripheral and central nervous systems, and this effect is likely mediated via modulation of oxidative stress, inflammation, and cell death pathways.
Collapse
Affiliation(s)
- Fatemeh Karami
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Jamaati
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Natalie Coleman-Fuller
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN, 55108, USA
| | - Maryam Shokrian Zeini
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - A Wallace Hayes
- University of South Florida College of Public Health and Institute for Integrative Toxicology, Michigan State University, East Lansing, USA
| | - Mina Gholami
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Salehirad
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Darabi
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Tosh N, Watson J, Lukas D, Tremewan R, Beard J, Galloway G, Haselhorst T, Young R, Crompton D, Mountford C. Two-dimensional correlated spectroscopy records reduced neurotransmission in blast-exposed artillery soldiers after live fire training. NMR IN BIOMEDICINE 2023:e4934. [PMID: 36940008 DOI: 10.1002/nbm.4934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/17/2023] [Accepted: 03/12/2023] [Indexed: 06/18/2023]
Abstract
There is a requirement for an objective method to determine a safe level of low-level military occupational blast, having recognised it can lead to neurological damage. The purpose of the current study was to evaluate the effect of artillery firing training on the neurochemistry of frontline soldiers using two-dimensional (2D) COrrelated SpectroscopY (2D COSY) in a 3-T clinical MR scanner. Ten men considered to be of sound health were evaluated before and after a week-long live firing exercise in two ways. Prior to the live fire exercise, all participants were screened by a clinical psychologist using a combination of clinical interviews and psychometric tests, and were then scanned with 3-T MRI. The protocols included T1- and T2-weighted images for diagnostic reporting and anatomical localisation and 2D COSY to record any neurochemical effects from the firing. No changes to the structural MRI were recorded. Nine substantive and statistically significant changes in the neurochemistry were recorded as a consequence of firing training. Glutamine and glutamate, glutathione, and two of the seven fucose-α (1-2)-glycans were significantly increased. N-acetyl aspartate, myo-inositol + creatine, and glycerol were also increased. Significant decreases were recorded for the glutathione cysteine moiety and tentatively assigned glycan with a 1-6 linkage (F2: 4.00, F1: 1.31 ppm). These molecules are part of three neurochemical pathways at the terminus of the neurons providing evidence of early markers of disruption to neurotransmission. Using this technology, the extent of deregulation can now be monitored for each frontline defender on a personalised basis. The capacity to monitor early a disruption in neurotransmitters, using the 2D COSY protocol, can observe the effect of firing and may be used to prevent or limit these events.
Collapse
Affiliation(s)
- Nathan Tosh
- Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Translational Research Institute, Woolloongabba, Queensland, Australia
- Radiology Department, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Julia Watson
- Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Translational Research Institute, Woolloongabba, Queensland, Australia
- Radiology Department, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Darren Lukas
- Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Rosanna Tremewan
- Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Jason Beard
- Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Graham Galloway
- Translational Research Institute, Woolloongabba, Queensland, Australia
| | | | - Ross Young
- Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - David Crompton
- Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Glycomics Institute, Southport, Queensland, Australia
| | - Carolyn Mountford
- Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Translational Research Institute, Woolloongabba, Queensland, Australia
- Radiology Department, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
- Glycomics Institute, Southport, Queensland, Australia
| |
Collapse
|
25
|
Vašková J, Kočan L, Vaško L, Perjési P. Glutathione-Related Enzymes and Proteins: A Review. Molecules 2023; 28:molecules28031447. [PMID: 36771108 PMCID: PMC9919958 DOI: 10.3390/molecules28031447] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The tripeptide glutathione is found in all eukaryotic cells, and due to the compartmentalization of biochemical processes, its synthesis takes place exclusively in the cytosol. At the same time, its functions depend on its transport to/from organelles and interorgan transport, in which the liver plays a central role. Glutathione is determined as a marker of the redox state in many diseases, aging processes, and cell death resulting from its properties and reactivity. It also uses other enzymes and proteins, which enables it to engage and regulate various cell functions. This paper approximates the role of these systems in redox and detoxification reactions such as conjugation reactions of glutathione-S-transferases, glyoxylases, reduction of peroxides through thiol peroxidases (glutathione peroxidases, peroxiredoxins) and thiol-disulfide exchange reactions catalyzed by glutaredoxins.
Collapse
Affiliation(s)
- Janka Vašková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 040 11 Košice, Slovakia
- Correspondence: (J.V.); (P.P.); Tel.: +42-155-234-3232 (J.V.)
| | - Ladislav Kočan
- Clinic of Anaesthesiology and Intensive Care Medicine, East Slovak Institute of Cardiovascular Disease, 040 11 Košice, Slovakia
| | - Ladislav Vaško
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 040 11 Košice, Slovakia
| | - Pál Perjési
- Institute of Pharmaceutical Chemistry, University of Pécs, 7600 Pécs, Hungary
- Correspondence: (J.V.); (P.P.); Tel.: +42-155-234-3232 (J.V.)
| |
Collapse
|
26
|
Ghanaatfar F, Ghanaatfar A, Isapour P, Farokhi N, Bozorgniahosseini S, Javadi M, Gholami M, Ulloa L, Coleman-Fuller N, Motaghinejad M. Is lithium neuroprotective? An updated mechanistic illustrated review. Fundam Clin Pharmacol 2023; 37:4-30. [PMID: 35996185 DOI: 10.1111/fcp.12826] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 06/17/2022] [Accepted: 08/19/2022] [Indexed: 01/25/2023]
Abstract
Neurodegeneration is a pathological process characterized by progressive neuronal impairment, dysfunction, and loss due to mitochondrial dysfunction, oxidative stress, inflammation, and apoptosis. Many studies have shown that lithium protects against neurodegeneration. Herein, we summarize recent clinical and laboratory studies on the neuroprotective effects of lithium against neurodegeneration and its potential to modulate mitochondrial dysfunction, oxidative stress, inflammation, and apoptosis. Recent findings indicate that lithium regulates critical intracellular pathways such as phosphatidylinositol-3 (PI3)/protein kinase B (Akt)/glycogen synthase kinase-3 (GSK3β) and PI3/Akt/response element-binding protein (CREB)/brain-derived neurotrophic factor (BDNF). We queried PubMed, Web of Science, Scopus, Elsevier, and other related databases using search terms related to lithium and its neuroprotective effect in various neurodegenerative diseases and events from January 2000 to May 2022. We reviewed the major findings and mechanisms proposed for the effects of lithium. Lithium's neuroprotective potential against neural cell degeneration is mediated by inducing anti-inflammatory factors, antioxidant enzymes, and free radical scavengers to prevent mitochondrial dysfunction. Lithium effects are regulated by two essential pathways: PI3/Akt/GSK3β and PI3/Akt/CREB/BDNF. Lithium acts as a neuroprotective agent against neurodegeneration by preventing inflammation, oxidative stress, apoptosis, and mitochondrial dysfunction using PI3/Akt/GSK3β and PI3/Akt/CREB/BDNF signaling pathways.
Collapse
Affiliation(s)
- Fateme Ghanaatfar
- Student Research Committee, School of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Ghanaatfar
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
| | - Parisa Isapour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Negin Farokhi
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | | | - Mahshid Javadi
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Gholami
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, North Carolina, USA
| | - Natalie Coleman-Fuller
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, USA
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Hughes KA, Misra B, Maghareh M, Bobbala S. Use of stimulatory responsive soft nanoparticles for intracellular drug delivery. NANO RESEARCH 2023; 16:6974-6990. [PMID: 36685637 PMCID: PMC9840428 DOI: 10.1007/s12274-022-5267-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 05/24/2023]
Abstract
Drug delivery has made tremendous advances in the last decade. Targeted therapies are increasingly common, with intracellular delivery highly impactful and sought after. Intracellular drug delivery systems have limitations due to imprecise and non-targeted release profiles. One way this can be addressed is through using stimuli-responsive soft nanoparticles, which contain materials with an organic backbone such as lipids and polymers. The choice of biomaterial is essential for soft nanoparticles to be responsive to internal or external stimuli. The nanoparticle must retain its integrity and payload in non-targeted physiological conditions while responding to particular intracellular environments where payload release is desired. Multiple internal and external factors could stimulate the intracellular release of drugs from nanoparticles. Internal stimuli include pH, oxidation, and enzymes, while external stimuli include ultrasound, light, electricity, and magnetic fields. Stimulatory responsive soft nanoparticulate systems specifically utilized to modulate intracellular delivery of drugs are explored in this review.
Collapse
Affiliation(s)
- Krystal A. Hughes
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Bishal Misra
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Maryam Maghareh
- Department of Clinical Pharmacy, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| |
Collapse
|
28
|
da Costa RO, Gadelha-Filho CVJ, de Aquino PEA, Lima LAR, de Lucena JD, Ribeiro WLC, Lima FAV, Neves KRT, de Barros Viana GS. Vitamin D (VD3) Intensifies the Effects of Exercise and Prevents Alterations of Behavior, Brain Oxidative Stress, and Neuroinflammation, in Hemiparkinsonian Rats. Neurochem Res 2023; 48:142-160. [PMID: 36028736 DOI: 10.1007/s11064-022-03728-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/31/2022] [Accepted: 08/15/2022] [Indexed: 01/11/2023]
Abstract
In the present study, we investigated the effects of physical exercise in the presence of Vitamin D3 (VD3), on 6-hydroxydopamine (6-OHDA)-lesioned hemiparkinsonian rats. The animals were divided into sham-operated (SO), 6-OHDA-lesioned, and 6-OHDA-lesioned plus VD3 (1 µg/kg, 21 days), in the absence (no exercise, NE) and presence (with exercise, WE) of physical exercise on a treadmill (30 min, speed of 20 cm/s, once a day/21 days). This procedure started, 24 h after the stereotaxic surgery (injections of 6-OHDA into the right striatum). The animals were then subjected to behavioral (rotarod, open field, and apomorphine tests) and their brain areas were dissected for neurochemical, dopamine (DA), 3,4-dihydroxyphenylacetic acid (DOPAC) determinations, and immunohistochemical studies for tyrosine hydroxylase (TH), dopamine transporter (DAT), and vitamin D receptor (VD3R). The effects on the brain oxidative stress: nitrite/nitrate, glutathione (GSH), and malondialdehyde (MDA) measurements were also evaluated. Behavioral changes of the 6-OHDA lesioned group were improved by exercise plus VD3. Similar results were observed in dopamine (DA) and 3,4-dihydroxyphenylacetic acid (DOPAC) concentrations increased by exercise and VD3, compared with SO groups. Additionally, tyrosine hydroxylase (TH) and dopamine transporter (DAT) immunoexpressions were decreased in the 6-OHDA-lesioned groups, with values normalized after exercise and VD3. The VD3 receptor immunoexpression decreased in the 6-OHDA (NE) group, and this was attenuated by exercise, especially after VD3. While 6-OHDA lesions increased, VD3 supplementation decreased the oxidative stress, which was intensified by exercise. VD3 showed neuroprotective properties that were intensified by physical exercise. These VD3 actions on hemiparkinsonian rats are possibly related to its antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Roberta Oliveira da Costa
- Graduate Program of Morphofunctional Sciences, Faculty of Medicine of the Federal University of Ceará, Fortaleza, Brazil
| | | | | | - Ludmila Araújo Rodrigues Lima
- Graduate Program of Morphofunctional Sciences, Faculty of Medicine of the Federal University of Ceará, Fortaleza, Brazil
| | - Jalles Dantas de Lucena
- Graduate Program of Morphofunctional Sciences, Faculty of Medicine of the Federal University of Ceará, Fortaleza, Brazil
| | | | | | - Kelly Rose Tavares Neves
- Graduate Program of Pharmacology, Faculty of Medicine of the Federal University of Ceará, Fortaleza, Brazil
| | - Glauce Socorro de Barros Viana
- Graduate Program of Morphofunctional Sciences, Faculty of Medicine of the Federal University of Ceará, Fortaleza, Brazil. .,Graduate Program of Pharmacology, Faculty of Medicine of the Federal University of Ceará, Fortaleza, Brazil.
| |
Collapse
|
29
|
Kinoshita C, Kubota N, Aoyama K. Glutathione Depletion and MicroRNA Dysregulation in Multiple System Atrophy: A Review. Int J Mol Sci 2022; 23:15076. [PMID: 36499400 PMCID: PMC9740333 DOI: 10.3390/ijms232315076] [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: 10/31/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022] Open
Abstract
Multiple system atrophy (MSA) is a rare neurodegenerative disease characterized by parkinsonism, cerebellar impairment, and autonomic failure. Although the causes of MSA onset and progression remain uncertain, its pathogenesis may involve oxidative stress via the generation of excess reactive oxygen species and/or destruction of the antioxidant system. One of the most powerful antioxidants is glutathione, which plays essential roles as an antioxidant enzyme cofactor, cysteine-storage molecule, major redox buffer, and neuromodulator, in addition to being a key antioxidant in the central nervous system. Glutathione levels are known to be reduced in neurodegenerative diseases. In addition, genes regulating redox states have been shown to be post-transcriptionally modified by microRNA (miRNA), one of the most important types of non-coding RNA. miRNAs have been reported to be dysregulated in several diseases, including MSA. In this review, we focused on the relation between glutathione deficiency, miRNA dysregulation and oxidative stress and their close relation with MSA pathology.
Collapse
Affiliation(s)
- Chisato Kinoshita
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Noriko Kubota
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
- Teikyo University Support Center for Women Physicians and Researchers, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Koji Aoyama
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| |
Collapse
|
30
|
Chen W, Liu H, Liu S, Kang Y, Nie Z, Lei H. Altered prefrontal neurochemistry in the DJ-1 knockout mouse model of Parkinson's disease: complementary semi-quantitative analyses with in vivo magnetic resonance spectroscopy and MALDI-MSI. Anal Bioanal Chem 2022; 414:7977-7987. [PMID: 36208327 DOI: 10.1007/s00216-022-04341-8] [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: 07/19/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022]
Abstract
In vivo proton magnetic resonance spectroscopy (1H-MRS) and matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) are two semi-quantitative analytical methods commonly used in neurochemical research. In this study, the two methods were used complementarily, in parallel, to investigate neurochemical perturbations in the medial prefrontal cortex (mPFC) of 9-month-old DJ-1 knockout mice, a well-established transgenic model for Parkinson's diseases. Convergingly, the results obtained with the two methods demonstrated that, compared with the wild-type (WT) mice, the DJ-1 knockout mice had significantly increased glutathione (GSH) level and GSH/glutamate (Glu) ratio in the mPFC, which likely presented an astrocytic compensatory mechanism in response to elevated regional oxidative stress induced by the loss of DJ-1 function. The results from this study also highlighted (1) the need to be cautious when interpreting the in vivo 1H-MRS results obtained from aged transgenic animals, in which the concentration of internal reference, being whether water or total creatine, could no longer be assumed to be the same as that in the age-matched WT animals, and (2) the necessity and importance of complementary analyses with more than one method under such circumstances.
Collapse
Affiliation(s)
- Wei Chen
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 30# Xiaohongshan West, Wuhan, 430071, Hubei, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Huihui Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Zhongguancun North First, Street 2, Beijing, 100190, China
| | - Sijie Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 30# Xiaohongshan West, Wuhan, 430071, Hubei, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Yan Kang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 30# Xiaohongshan West, Wuhan, 430071, Hubei, People's Republic of China
| | - Zongxiu Nie
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China. .,Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Zhongguancun North First, Street 2, Beijing, 100190, China.
| | - Hao Lei
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 30# Xiaohongshan West, Wuhan, 430071, Hubei, People's Republic of China. .,University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| |
Collapse
|
31
|
Al-Ghraiybah NF, Wang J, Alkhalifa AE, Roberts AB, Raj R, Yang E, Kaddoumi A. Glial Cell-Mediated Neuroinflammation in Alzheimer's Disease. Int J Mol Sci 2022; 23:10572. [PMID: 36142483 PMCID: PMC9502483 DOI: 10.3390/ijms231810572] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder; it is the most common cause of dementia and has no treatment. It is characterized by two pathological hallmarks, the extracellular deposits of amyloid beta (Aβ) and the intraneuronal deposits of Neurofibrillary tangles (NFTs). Yet, those two hallmarks do not explain the full pathology seen with AD, suggesting the involvement of other mechanisms. Neuroinflammation could offer another explanation for the progression of the disease. This review provides an overview of recent advances on the role of the immune cells' microglia and astrocytes in neuroinflammation. In AD, microglia and astrocytes become reactive by several mechanisms leading to the release of proinflammatory cytokines that cause further neuronal damage. We then provide updates on neuroinflammation diagnostic markers and investigational therapeutics currently in clinical trials to target neuroinflammation.
Collapse
Affiliation(s)
- Nour F. Al-Ghraiybah
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Junwei Wang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Amer E. Alkhalifa
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Andrew B. Roberts
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Ruchika Raj
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Euitaek Yang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| |
Collapse
|
32
|
Choi IY, Taylor MK, Lee P, Alhayek SA, Bechtel M, Hamilton-Reeves J, Spaeth K, Adany P, Sullivan DK. Milk intake enhances cerebral antioxidant (glutathione) concentration in older adults: A randomized controlled intervention study. Front Nutr 2022; 9:811650. [PMID: 36046132 PMCID: PMC9421260 DOI: 10.3389/fnut.2022.811650] [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: 11/09/2021] [Accepted: 07/18/2022] [Indexed: 11/25/2022] Open
Abstract
Background A major antioxidant, glutathione (GSH), is a key factor in the antioxidant defense mechanism against oxidative stress and aging-related functional declines. Our previous observational study showed positive correlations between brain GSH concentrations and dairy food consumption, particularly milk (p < 0.001), in older adults. Objective To investigate whether a recommended amount of milk intake (3 cups/day) in low dairy consumers enhances brain GSH concentrations through an intervention trial. Methods Seventy-three older adults (60–89 years) with a low dairy intake (≤1.5 servings/day) were randomized (5:2 ratio) in this 3-month randomized clinical trial. The intervention group was provided 1% milk weekly and instructed to consume 3 cups of milk/day for 3 months while the control group continued their habitual intake of total dairy ≤ 1.5 servings/day (<1 cup of milk/day). Brain GSH concentrations were measured in the fronto-parietal region using our unique 3 T magnetic resonance chemical shift imaging technique at baseline and 3 months. Results Among 73 randomized participants, 66 participants (49 intervention; 17 controls) completed the study. Milk intake in the intervention group increased from 0.2 ± 0.3 cups/day to 3.0 ± 0.6 cups/day (p < 0.001) between baseline and the end of the study, while milk intake in the control group did not differ throughout the study duration (0.4 ± 0.4 cups/day). The intervention group showed increases in brain GSH concentrations by 7.4 ± 11.7% (p < 0.001) in parietal and 4.7 ± 9.8% (p = 0.003) in fronto-parietal regions, and 4.6 ± 8.7% (p < 0.001) in overall brain concentration after the intervention compared with baseline, while the control group showed no changes. Conclusion This study provides evidence that milk serves as a good dietary source to increase and/or restore brain GSH concentrations in older adults. Identifying dietary sources that effectively enhance antioxidant defenses and neuroprotection could lead to the development of new strategies to promote brain health in the aging population. Clinical trial registration [https://ClinicalTrials.gov], identifier [NCT02957422].
Collapse
Affiliation(s)
- In-Young Choi
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States.,Hoglund Biomedical Imaging Center, University of Kansas Medical Center, Kansas City, KS, United States.,Department of Radiology, University of Kansas Medical Center, Kansas City, KS, United States.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Matthew K Taylor
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
| | - Phil Lee
- Hoglund Biomedical Imaging Center, University of Kansas Medical Center, Kansas City, KS, United States.,Department of Radiology, University of Kansas Medical Center, Kansas City, KS, United States.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Sibelle Alwatchi Alhayek
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
| | - Misty Bechtel
- Department of Urology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jill Hamilton-Reeves
- Department of Urology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Kendra Spaeth
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
| | - Peter Adany
- Hoglund Biomedical Imaging Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Debra K Sullivan
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
33
|
Junghans M, John F, Cihankaya H, Schliebs D, Winklhofer KF, Bader V, Matschke J, Theiss C, Matschke V. ROS scavengers decrease γH2ax spots in motor neuronal nuclei of ALS model mice in vitro. Front Cell Neurosci 2022; 16:963169. [PMID: 36119129 PMCID: PMC9470831 DOI: 10.3389/fncel.2022.963169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/08/2022] [Indexed: 12/05/2022] Open
Abstract
Background: Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disease characterized by the loss of motor neurons in cerebral cortex, brainstem and spinal cord. Numerous studies have demonstrated signs of oxidative stress in postmortem neuronal tissue, cerebrospinal fluid, plasma and urine of ALS patients, without focusing on the specific processes within motor neurons. Thus, we aimed to investigate the relevance of reactive oxygen species (ROS) detoxification mechanisms and its consequences on the formation of toxic/lethal DNA double strand breaks (DSBs) in the ALS model of the Wobbler mouse. Methods: Live cell imaging in dissociated motor neuronal cultures was used to investigate the production of ROS using Dihydroethidium (DHE). The expression levels of ROS detoxifying molecules were investigated by qPCR as well as Western blots. Furthermore, the expression levels of DNA damage response proteins p53bp1 and H2ax were investigated using qPCR and immunofluorescence staining. Proof-of-principle experiments using ROS scavengers were performed in vitro to decipher the influence of ROS on the formation of DNA double strand breaks quantifying the γH2ax spots formation. Results: Here, we verified an elevated ROS-level in spinal motor neurons of symptomatic Wobbler mice in vitro. As a result, an increased number of DNA damage response proteins p53bp1 and γH2ax in dissociated motor neurons of the spinal cord of Wobbler mice was observed. Furthermore, we found a significantly altered expression of several antioxidant molecules in the spinal cord of Wobbler mice, suggesting a deficit in ROS detoxification mechanisms. This hypothesis could be verified by using ROS scavenger molecules in vitro to reduce the number of γH2ax foci in dissociated motor neurons and thus counteract the harmful effects of ROS. Conclusion: Our data indicate that maintenance of redox homeostasis may play a key role in the therapy of the neurodegenerative disease ALS. Our results underline a necessity for multimodal treatment approaches to prolong the average lifespan of motor neurons and thus slow down the progression of the disease, since a focused intervention in one pathomechanism seems to be insufficient in ALS therapy.
Collapse
Affiliation(s)
- Maya Junghans
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Felix John
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Hilal Cihankaya
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Daniel Schliebs
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Konstanze F. Winklhofer
- Department of Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Cluster of Excellence RESOLV, Bochum, Germany
| | - Verian Bader
- Department of Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Department of Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Johann Matschke
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
- *Correspondence: Veronika Matschke
| |
Collapse
|
34
|
Dipeptide of ψ-GSH Inhibits Oxidative Stress and Neuroinflammation in an Alzheimer's Disease Mouse Model. Antioxidants (Basel) 2022; 11:antiox11061075. [PMID: 35739972 PMCID: PMC9219802 DOI: 10.3390/antiox11061075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/10/2022] Open
Abstract
Supplementation of glutathione (GSH) levels through varying formulations or precursors has thus far appeared to be a tenable strategy to ameliorate disease-associated oxidative stress. Metabolic liability of GSH and its precursors, i.e., hydrolysis by the ubiquitous γ-glutamyl transpeptidase (γ-GT), has limited successful clinical translation due to poor bioavailability. We addressed this problem through the design of γ-GT-resistant GSH analogue, ψ-GSH, which successfully substituted in GSH-dependent enzymatic systems and also offered promise as a therapeutic for Alzheimer's disease (AD). With the aim to improve its bioavailability, we studied the utility of a ψ-GSH precursor, dipeptide 2, as a potential AD therapeutic. Compound 2 retains the γ-GT stable ureide linkage and the thiol group for antioxidant property. By engaging glutathione synthetase, compound 2 was able to generate ψ-GSH in vivo. It was found to be a modest cofactor of glutathione peroxidase and prevented cytotoxicity of Aβ1-42-aggregates in vitro. Studies of compound 2 in an acute AD model generated by intracerebroventricular injection of Aβ1-42 showed cognitive benefits, which were augmented by its combination with glycine along with mitigation of oxidative stress and inflammatory pathology. Collectively, these results support further optimization and evaluation of ψ-GSH dipeptide as a potential therapeutic in transgenic AD models.
Collapse
|
35
|
Zhao X, He Y, Zhang Y, Wan H, Wan H, Yang J. Inhibition of Oxidative Stress: An Important Molecular Mechanism of Chinese Herbal Medicine (Astragalus membranaceus, Carthamus tinctorius L., Radix Salvia Miltiorrhizae, etc.) in the Treatment of Ischemic Stroke by Regulating the Antioxidant System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1425369. [PMID: 35651725 PMCID: PMC9151006 DOI: 10.1155/2022/1425369] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/11/2022] [Indexed: 01/07/2023]
Abstract
Ischemic stroke is a severe cerebrovascular disease with high mortality and morbidity. Traditional Chinese medicine (TCM) has been utilized for thousands of years in China and is becoming increasingly popular all over the world, especially for the treatments of ischemic stroke. More and more evidences have implicated that oxidative stress has been closely related with ischemic stroke. This review will concentrate on the evidence of the action mechanism of Chinese herbal medicine and its active ingredient in preventing ischemic stroke by modulating redox signaling and oxidative stress pathways and providing references for clinical treatment and scientific research applications.
Collapse
Affiliation(s)
- Xixi Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yangyang Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Haofang Wan
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Haitong Wan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiehong Yang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
36
|
Watson J, Lukas D, Vickers ER, Galloway G, Mountford CE. Case Report: Capacity to Objectively Monitor the Response of a Chronic Pain Patient to Treatment. FRONTIERS IN NEUROIMAGING 2022; 1:831216. [PMID: 37555159 PMCID: PMC10406213 DOI: 10.3389/fnimg.2022.831216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/29/2022] [Indexed: 08/10/2023]
Abstract
Response to pain therapy is currently by patient self-report. We demonstrate that by evaluating the neurochemistry of a patient, using two-dimensional Correlated SpectroscopY (2D COSY) in a 3T MRI scanner, response to therapy can be recorded. A chronic temporomandibular joint (TMJ) pain patient was evaluated by a pain physician specializing in temporomandibular disorders (TMD), and by 2D COSY, before, and 6 days after treatment with Botulinum Toxin A. Prior to treatment the self-reported pain score was 8/10 and reduced to 0/10 within 24 h of treatment. The neurochemistry of the patient prior to treatment was typical of chronic pain. In particular, the Fuc-α(1-2) glycans were affected. Following treatment, the substrates, α-L Fucose, were elevated and the Fuc-α(1-2) glycans repopulated. The depletion of the molecule assigned the glutathione cysteine moiety, with chronic pain, is indicative of a Glutathione redox imbalance linked to neurodegeneration. This new approach to monitor pain could help discriminate the relative contributions in the complex interplay of the sensory and affective (emotional suffering) components of pain leading to appropriate individualized pharmaceutical drug regimens.
Collapse
Affiliation(s)
- Julia Watson
- Faculty of Health, School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- Princess Alexandra Hospital, Department of Radiology, Woolloongabba, QLD, Australia
- Department of Imaging Technology, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Darren Lukas
- Institute for Glycomics, Gold Coast Campus, Griffith University, Southport, QLD, Australia
| | | | - Graham Galloway
- Faculty of Health, School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- Department of Imaging Technology, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Carolyn E. Mountford
- Faculty of Health, School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- Princess Alexandra Hospital, Department of Radiology, Woolloongabba, QLD, Australia
- Department of Imaging Technology, Translational Research Institute, Woolloongabba, QLD, Australia
- Institute for Glycomics, Gold Coast Campus, Griffith University, Southport, QLD, Australia
| |
Collapse
|
37
|
Koszła O, Sołek P, Kędzierska E, Listos P, Castro M, Kaczor AA. In vitro and in vivo evaluation of antioxidant and neuroprotective properties of antipsychotic D2AAK1. Neurochem Res 2022; 47:1778-1789. [PMID: 35380398 DOI: 10.1007/s11064-022-03570-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 12/30/2022]
Abstract
The susceptibility of neurons to free radical toxicity partially underlies the pathomechanism of neurodegenerative diseases. On the other hand, excitotoxicity also contributes to neurodegeneration. Our previous studies demonstrated the unique properties of D2AAK1 as a potent multi-target ligand of aminergic G protein-coupled receptors (GPCRs) which dose-dependently stimulates growth, survival of neurons, and promotes their integrity. The aim of our study was to investigate the potential neuroprotective and antioxidant properties of D2AAK1. Here we show that D2AAK1 activates cellular and molecular neuroprotective mechanisms, prevents cells from excitotoxicity and free radicals. Furthermore, D2AAK1 induced no genotoxic events in neuronal cells in vitro. Most importantly, D2AAK1 protects neurons from the effects of high temperatures by molecular chaperones activation. The D2AAK1 effects on selected organs was further evaluated in mice and no pathological changes were observed after chronic administration. In the light of our experiments, D2AAK1 can be further developed into a potential treatment for neurodegenerative diseases, in particular related to memory impairment. In summary, D2AAK1 has promising properties for potential treatments of neurodegenerative diseases.
Collapse
Affiliation(s)
- Oliwia Koszła
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St, 20-093, Lublin, Poland.
| | - Przemysław Sołek
- Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, 1 Pigonia St, 35-310, Rzeszow, Poland
| | - Ewa Kędzierska
- Department of Pharmacology and Pharmacodynamics, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St, 20-093, Lublin, Poland
| | - Piotr Listos
- Department of Pathomorphology and Forensic Veterinary Medicine, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 30 Głęboka St, 20-033, Lublin, Poland
| | - Marián Castro
- Department of Pharmacology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Avda de Barcelona, E-15782, Santiago de Compostela, Spain
| | - Agnieszka A Kaczor
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St, 20-093, Lublin, Poland. .,School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
38
|
α-Lipoic Acid Strengthens the Antioxidant Barrier and Reduces Oxidative, Nitrosative, and Glycative Damage, as well as Inhibits Inflammation and Apoptosis in the Hypothalamus but Not in the Cerebral Cortex of Insulin-Resistant Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7450514. [PMID: 35391928 PMCID: PMC8983239 DOI: 10.1155/2022/7450514] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/24/2022] [Accepted: 03/18/2022] [Indexed: 12/17/2022]
Abstract
The research determined the role of α-lipoic acid (ALA) in reducing the brain manifestations of insulin resistance. The mechanism of ALA action is mainly based on its ability to “scavenge” oxygen free radicals and stimulate biosynthesis of reduced glutathione (GSH), considered the most critical brain antioxidant. Although the protective effect of ALA is widely documented in various diseases, there are still no studies assessing the influence of ALA on brain metabolism in the context of insulin resistance and type 2 diabetes. The experiment was conducted on male Wistar rats fed a high-fat diet for ten weeks with intragastric administration of ALA for four weeks. We are the first to demonstrate that ALA improves the function of enzymatic and nonenzymatic brain antioxidant systems, but the protective effects of ALA were mainly observed in the hypothalamus of insulin-resistant rats. Indeed, ALA caused a significant increase in superoxide dismutase, catalase, peroxidase, and glutathione reductase activities, as well as GSH concentration and redox potential ([GSH]2/[GSSG]) in the hypothalamus of HFD-fed rats. A consequence of antioxidant barrier enhancement by ALA is the reduction of oxidation, glycation, and nitration of brain proteins, lipids, and DNA. The protective effects of ALA result from hypothalamic activation of the transcription factor Nrf2 and inhibition of NF-κB. In the hypothalamus of insulin-resistant rats, we demonstrated reduced levels of oxidation (AOPP) and glycation (AGE) protein products, 4-hydroxynoneal, 8-isoprostanes, and 3-nitrotyrosine and, in the cerebral cortex, lower levels of 8-hydroxydeoxyguanosine and peroxynitrite. In addition, we demonstrated that ALA decreases levels of proinflammatory TNF-α but also increases the synthesis of anti-inflammatory IL-10 in the hypothalamus of insulin-resistant rats. ALA also prevents neuronal apoptosis, confirming its multidirectional effects within the brain. Interestingly, we have shown no correlation between brain and serum/plasma oxidative stress biomarkers, indicating the different nature of redox imbalance at the central and systemic levels. To summarize, ALA improves antioxidant balance and diminishes oxidative/glycative stress, protein nitrosative damage, inflammation, and apoptosis, mainly in the hypothalamus of insulin-resistant rats. Further studies are needed to determine the molecular mechanism of ALA action within the brain.
Collapse
|
39
|
The Influence of Intracellular Glutathione Levels on the Induction of Nrf2-Mediated Gene Expression by α-Dicarbonyl Precursors of Advanced Glycation End Products. Nutrients 2022; 14:nu14071364. [PMID: 35405976 PMCID: PMC9003139 DOI: 10.3390/nu14071364] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 12/03/2022] Open
Abstract
α-Dicarbonyl compounds, particularly methylglyoxal (MGO), glyoxal (GO), and 3-deoxyglucosone (3-DG), are highly reactive precursors for the formation of advanced glycation end products (AGEs). They are formed in vivo and during food processing. This study aimed to investigate the role of intracellular glutathione (GSH) levels in the induction of Nrf2-mediated gene expression by α-dicarbonyl compounds. The reactions between α-dicarbonyl compounds (MGO, GO, and 3-DG) and GSH were studied by LC-MS in a cell-free system. It was shown that these three α-dicarbonyl compounds react instantaneously with GSH, with the GSH-mediated scavenging decreasing in the order MGO > GO > 3DG. Furthermore, in a cell-based reporter gene assay MGO, GO, and 3-DG were able to induce Nrf2-mediated gene expression in a dose-dependent manner. Modulation of intracellular GSH levels showed that the cytotoxicity and induction of the Nrf2-mediated pathway by MGO, GO and 3-DG was significantly enhanced by depletion of GSH, while a decrease in Nrf2-activation by MGO and GO but not 3-DG was observed upon an increase of the cellular GSH levels. Our results reveal subtle differences in the role of GSH in protection against the three typical α-dicarbonyl compounds and in their induction of Nrf2-mediated gene expression, and point at a dual biological effect of the α-dicarbonyl compounds, being reactive toxic electrophiles and -as a consequence- able to induce Nrf2-mediated protective gene expression, with MGO being most reactive.
Collapse
|
40
|
Kim D, Kim S, Sung A, Patel N, Wong N, Conboy MJ, Conboy IM. Autologous treatment for ALS with implication for broad neuroprotection. Transl Neurodegener 2022; 11:16. [PMID: 35272709 PMCID: PMC8915496 DOI: 10.1186/s40035-022-00290-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/18/2022] [Indexed: 01/20/2023] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is characterized by a progressive loss of motor neurons (MNs), leading to paralysis, respiratory failure and death within 2–5 years of diagnosis. The exact mechanisms of sporadic ALS, which comprises 90% of all cases, remain unknown. In familial ALS, mutations in superoxide dismutase (SOD1) cause 10% of cases. Methods ALS patient-derived human-induced pluripotent stem cells (ALS hiPSCs, harboring the SOD1AV4 mutation), were differentiated to MNs (ALS-MNs). The neuroprotective effects of conditioned medium (CM) of hESCs (H9), wt hiPSCs (WTC-11) and the ALS iPSCs, on MN apoptosis and viability, formation and maintenance of neurites, mitochondrial activity and expression of inflammatory genes, were examined. For in vivo studies, 200 μl of CM from the ALS iPSCs (CS07 and CS053) was injected subcutaneously into the ALS model mice (transgenic for the human SOD1G93A mutation). Animal agility and strength, muscle innervation and mass, neurological score, onset of paralysis and lifespan of the ALS mice were assayed. After observing significant disease-modifying effects, the CM was characterized biochemically by fractionation, comparative proteomics, and epigenetic screens for the dependence on pluripotency. CM of fibroblasts that were differentiated from the wt hiPSCs lacked any neuroprotective activity and was used as a negative control throughout the studies. Results The secretome of PSCs including the ALS patient iPSCs was neuroprotective in the H2O2 model. In the model with pathogenic SOD1 mutation, ALS iPSC-CM attenuated all examined hallmarks of ALS pathology, rescued human ALS-MNs from denervation and death, restored mitochondrial health, and reduced the expression of inflammatory genes. The ALS iPSC-CM also improved neuro-muscular health and function, and delayed paralysis and morbidity in ALS mice. Compared side by side, cyclosporine (CsA), a mitochondrial membrane blocker that prevents the leakage of mitochondrial DNA, failed to avert the death of ALS-MNs, although CsA and ALS iPSC-CM equally stabilized MN mitochondria and attenuated inflammatory genes. Biochemical characterization, comparative proteomics, and epigenetic screen all suggested that it was the interactome of several key proteins from different fractions of PSC-CM that delivered the multifaceted neuroprotection. Conclusions This work introduces and mechanistically characterizes a new biologic for treating ALS and other complex neurodegenerative diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s40035-022-00290-5.
Collapse
Affiliation(s)
- Daehwan Kim
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Subin Kim
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Ashley Sung
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Neetika Patel
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Nathan Wong
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Michael J Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
41
|
Haider M, Elsherbeny A, Pittalà V, Consoli V, Alghamdi MA, Hussain Z, Khoder G, Greish K. Nanomedicine Strategies for Management of Drug Resistance in Lung Cancer. Int J Mol Sci 2022; 23:1853. [PMID: 35163777 PMCID: PMC8836587 DOI: 10.3390/ijms23031853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/01/2022] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Lung cancer (LC) is one of the leading causes of cancer occurrence and mortality worldwide. Treatment of patients with advanced and metastatic LC presents a significant challenge, as malignant cells use different mechanisms to resist chemotherapy. Drug resistance (DR) is a complex process that occurs due to a variety of genetic and acquired factors. Identifying the mechanisms underlying DR in LC patients and possible therapeutic alternatives for more efficient therapy is a central goal of LC research. Advances in nanotechnology resulted in the development of targeted and multifunctional nanoscale drug constructs. The possible modulation of the components of nanomedicine, their surface functionalization, and the encapsulation of various active therapeutics provide promising tools to bypass crucial biological barriers. These attributes enhance the delivery of multiple therapeutic agents directly to the tumor microenvironment (TME), resulting in reversal of LC resistance to anticancer treatment. This review provides a broad framework for understanding the different molecular mechanisms of DR in lung cancer, presents novel nanomedicine therapeutics aimed at improving the efficacy of treatment of various forms of resistant LC; outlines current challenges in using nanotechnology for reversing DR; and discusses the future directions for the clinical application of nanomedicine in the management of LC resistance.
Collapse
Affiliation(s)
- Mohamed Haider
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (Z.H.); (G.K.)
| | - Amr Elsherbeny
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Valeria Pittalà
- Department of Drug and Health Science, University of Catania, 95125 Catania, Italy; (V.P.); (V.C.)
| | - Valeria Consoli
- Department of Drug and Health Science, University of Catania, 95125 Catania, Italy; (V.P.); (V.C.)
| | - Maha Ali Alghamdi
- Department of Biotechnology, College of Science, Taif University, Taif 21974, Saudi Arabia;
- Department of Molecular Medicine, Princess Al-Jawhara Centre for Molecular Medicine, School of Medicine and Medical Sciences, Arabian Gulf University, Manama 329, Bahrain;
| | - Zahid Hussain
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (Z.H.); (G.K.)
| | - Ghalia Khoder
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (Z.H.); (G.K.)
| | - Khaled Greish
- Department of Molecular Medicine, Princess Al-Jawhara Centre for Molecular Medicine, School of Medicine and Medical Sciences, Arabian Gulf University, Manama 329, Bahrain;
| |
Collapse
|
42
|
Chen JJ, Thiyagarajah M, Song J, Chen C, Herrmann N, Gallagher D, Rapoport MJ, Black SE, Ramirez J, Andreazza AC, Oh P, Marzolini S, Graham SJ, Lanctôt KL. Altered central and blood glutathione in Alzheimer's disease and mild cognitive impairment: a meta-analysis. Alzheimers Res Ther 2022; 14:23. [PMID: 35123548 PMCID: PMC8818133 DOI: 10.1186/s13195-022-00961-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 01/06/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND Increasing evidence implicates oxidative stress (OS) in Alzheimer disease (AD) and mild cognitive impairment (MCI). Depletion of the brain antioxidant glutathione (GSH) may be important in OS-mediated neurodegeneration, though studies of post-mortem brain GSH changes in AD have been inconclusive. Recent in vivo measurements of the brain and blood GSH may shed light on GSH changes earlier in the disease. AIM To quantitatively review in vivo GSH in AD and MCI compared to healthy controls (HC) using meta-analyses. METHOD Studies with in vivo brain or blood GSH levels in MCI or AD with a HC group were identified using MEDLINE, PsychInfo, and Embase (1947-June 2020). Standardized mean differences (SMD) and 95% confidence intervals (CI) were calculated for outcomes using random effects models. Outcome measures included brain GSH (Meshcher-Garwood Point Resolved Spectroscopy (MEGA-PRESS) versus non-MEGA-PRESS) and blood GSH (intracellular versus extracellular) in AD and MCI. The Q statistic and Egger's test were used to assess heterogeneity and risk of publication bias, respectively. RESULTS For brain GSH, 4 AD (AD=135, HC=223) and 4 MCI (MCI=213, HC=211) studies were included. For blood GSH, 26 AD (AD=1203, HC=1135) and 7 MCI (MCI=434, HC=408) studies were included. Brain GSH overall did not differ in AD or MCI compared to HC; however, the subgroup of studies using MEGA-PRESS reported lower brain GSH in AD (SMD [95%CI] -1.45 [-1.83, -1.06], p<0.001) and MCI (-1.15 [-1.71, -0.59], z=4.0, p<0.001). AD had lower intracellular and extracellular blood GSH overall (-0.87 [-1. 30, -0.44], z=3.96, p<0.001). In a subgroup analysis, intracellular GSH was lower in MCI (-0.66 [-1.11, -0.21], p=0.025). Heterogeneity was observed throughout (I2 >85%) and not fully accounted by subgroup analysis. Egger's test indicated risk of publication bias. CONCLUSION Blood intracellular GSH decrease is seen in MCI, while both intra- and extracellular decreases were seen in AD. Brain GSH is decreased in AD and MCI in subgroup analysis. Potential bias and heterogeneity suggest the need for measurement standardization and additional studies to explore sources of heterogeneity.
Collapse
Affiliation(s)
- Jinghan Jenny Chen
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Room FG52, Toronto, ON, M4N 3M5, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Mathura Thiyagarajah
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Room FG52, Toronto, ON, M4N 3M5, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Jianmeng Song
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Room FG52, Toronto, ON, M4N 3M5, Canada
| | - Clara Chen
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Room FG52, Toronto, ON, M4N 3M5, Canada
| | - Nathan Herrmann
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Room FG52, Toronto, ON, M4N 3M5, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Geriatric Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Damien Gallagher
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Geriatric Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Mark J Rapoport
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Geriatric Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Sandra E Black
- Evaluative Clinical Sciences, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
- KITE-Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| | - Joel Ramirez
- Evaluative Clinical Sciences, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Ana C Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Paul Oh
- KITE-Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| | - Susan Marzolini
- KITE-Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| | - Simon J Graham
- Physical Sciences, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Krista L Lanctôt
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Room FG52, Toronto, ON, M4N 3M5, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Geriatric Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
- Evaluative Clinical Sciences, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.
| |
Collapse
|
43
|
Goel Y, Fouda R, Gupta K. Endoplasmic Reticulum Stress in Chemotherapy-Induced Peripheral Neuropathy: Emerging Role of Phytochemicals. Antioxidants (Basel) 2022; 11:antiox11020265. [PMID: 35204148 PMCID: PMC8868275 DOI: 10.3390/antiox11020265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/22/2022] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a significant dose-limiting long-term sequela in cancer patients undergoing treatment, often leading to discontinuation of treatment. No established therapy exists to prevent and/or ameliorate CIPN. Reactive oxygen species (ROS) and mitochondrial dysregulation have been proposed to underlie the pathobiology of CIPN. However, interventions to prevent and treat CIPN are largely ineffective. Additional factors and mechanism-based targets need to be identified to develop novel strategies to target CIPN. The role of oxidative stress appears to be central, but the contribution of endoplasmic reticulum (ER) stress remains under-examined in the pathobiology of CIPN. This review describes the significance of ER stress and its contribution to CIPN, the protective role of herbal agents in countering ER stress in nervous system-associated disorders, and their possible repurposing for preventing CIPN.
Collapse
Affiliation(s)
- Yugal Goel
- Hematology/Oncology, Department of Medicine, University of California, Irvine, CA 92697, USA; (Y.G.); (R.F.)
| | - Raghda Fouda
- Hematology/Oncology, Department of Medicine, University of California, Irvine, CA 92697, USA; (Y.G.); (R.F.)
| | - Kalpna Gupta
- Hematology/Oncology, Department of Medicine, University of California, Irvine, CA 92697, USA; (Y.G.); (R.F.)
- VA Medical Center, Southern California Institute for Research and Education, Long Beach, CA 90822, USA
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
- Correspondence:
| |
Collapse
|
44
|
Egba SI, Famurewa AC, Omoruyi LE. Buchholzia coriacea seed extract attenuates mercury-induced cerebral and cerebellar oxidative neurotoxicity via NO signaling and suppression of oxidative stress, adenosine deaminase and acetylcholinesterase activities in rats. AVICENNA JOURNAL OF PHYTOMEDICINE 2022; 12:42-53. [PMID: 35145894 PMCID: PMC8801217 DOI: 10.22038/ajp.2021.18262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Mercury (Hg) is a classic cumulative neurotoxicant implicated in neuronal deficit via oxidative damage and inflammatory responses. We sought to investigate whether Buccholzia coriacea seed methanol extract (BCSE) would modulate oxidative neurotoxicity induced by Hg in rats. MATERIALS AND METHODS Rats were orally treated with BCSE (200 or 400 mg/kg body weight of rat) for 28 days, while Hg was administered from day 15 to day 28. After sacrifice, antioxidant enzyme activities, reduced glutathione (GSH), nitric oxide (NO), malondialdehyde (MDA), and acetylcholinesterase (AchE) and adenine deaminase (ADA) activities were evaluated in the cerebrum and cerebellum of rats. RESULTS Mercury induced significant depressions in catalase (CAT) and glutathione peroxidase (GPx) activities and GSH levels, whereas levels of NO and activities of AchE and ADA markedly increased. The histopathology of the brain tissues confirmed these changes. In contrast, BCSE administration prominently modulated the brain NO production and reversed the Hg-induced biochemical alterations comparable to normal control. CONCLUSION Methanol extract of B. coriacea seeds protects the cerebrum and cerebellum against Hg-induced brain damage via its antioxidant and NO modulatory actions.
Collapse
Affiliation(s)
- Simeon I Egba
- Department of Biochemistry, Michael Okpara University of Agriculture, Umudike, Abia State, Nigeria
| | - Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Alex-Ekwueme Federal University, Ndufu-Alike, Ikwo, Ebonyi State, Nigeria
| | - Lilian E Omoruyi
- Department of Biochemistry, Michael Okpara University of Agriculture, Umudike, Abia State, Nigeria
| |
Collapse
|
45
|
Tripathi AS, Bansod P, Swathi KP. Activation of 5-HT 1b/d receptor restores the cognitive function by reducing glutamate release, deposition of β-amyloid and TLR-4 pathway in the brain of scopolamine-induced dementia in rat. J Pharm Pharmacol 2021; 73:1592-1598. [PMID: 34244776 DOI: 10.1093/jpp/rgab095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 06/07/2021] [Indexed: 11/13/2022]
Abstract
OBJECTIVES This study evaluates the effect of 5-HT 1b/d agonist on cognitive function in scopolamine (SPN)-induced dementia in the rat. METHODS Dementia was induced by administration of SPN 2 mg/kg/day, intraperitoneally, for a duration of 21 days. The effect of zolmitriptan (ZMT) 30 mg/kg, intraperitoneally, was observed on cognitive function, and the parameters of oxidative stress like malondialdehyde (MDA) level, nitric oxide (NO), superoxide dismutase (SOD) and glutathione peroxidase (GPX) were estimated at the end. Histopathology study of brain tissue was performed for the determination of β-amyloid peptide, and qRT-PCR was used to determine the mRNA expression of Toll-like receptor 4 (TLR-4), IL-17 and β-amyloid. KEY FINDINGS Data of the study suggested that treatment with ZMT alone and in combination with DMP (dextromethorphan) significantly (P < 0.01) decreases the escape latency in conditioned avoidance response (CAR) and transfer latency in elevated plus maze (EPM) as compared with negative control group. Moreover, the result of Morris water maze (MWM) shows an increase in retention time and a decrease in escape latency in ZMT alone and in combination with DMP-treated group of SPN-induced dementia than in the negative control group. There was a significant decrease in MDA and NO and increase in SOD and GPX in the brain tissues of ZMT and ZMT + DMP-treated group than negative control group. Histopathology study also suggested that the concentration of Aβ peptide decreases in the brain tissues in ZMT and ZMT + DMP-treated group than the negative control group. Moreover, ZMT treatment ameliorates the altered mRNA expression of TLR-4 and IL-17 in the brain tissue of SPN-induced dementia rat. CONCLUSIONS In conclusion, ZMT restores the cognitive functions and impaired memory in SPN-induced dementia in the rat by decreasing oxidative stress and Aβ peptide in the brain tissue of rat.
Collapse
Affiliation(s)
- Alok Shiomurti Tripathi
- Amity Institute of Pharmacy, Amity University, Sector 125, Noida, Uttar Pradesh, India
- Department of Pharmacology, P. Wadhwani College of Pharmacy, Yavatmal, Maharashtra, India
| | - Prajakta Bansod
- Department of Pharmacology, P. Wadhwani College of Pharmacy, Yavatmal, Maharashtra, India
| | - K P Swathi
- Department of Pharmacology, College of Pharmacy, Kannur Medical College, Kannur, Kerala, India
| |
Collapse
|
46
|
Christopher Kwon YI, Xie W, Zhu H, Xie J, Shinn K, Juckel N, Vince R, More SS, Lee MK. γ-Glutamyl-Transpeptidase-Resistant Glutathione Analog Attenuates Progression of Alzheimer's Disease-like Pathology and Neurodegeneration in a Mouse Model. Antioxidants (Basel) 2021; 10:antiox10111796. [PMID: 34829667 PMCID: PMC8614797 DOI: 10.3390/antiox10111796] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/26/2021] [Accepted: 11/08/2021] [Indexed: 02/01/2023] Open
Abstract
Oxidative stress in Alzheimer’s disease (AD) is mediated, in part, by the loss of glutathione (GSH). Previous studies show that γ-glutamyl transpeptidase (GGT)-resistant GSH analog, Ψ-GSH, improves brain GSH levels, reduces oxidative stress markers in brains of APP/PS1 transgenic mice, a mouse model of AD, and attenuates early memory deficits in the APP/PS1 model. Herein, we examined whether Ψ-GSH can attenuate the disease progression when administered following the onset of AD-like pathology in vivo. Cohorts of APP/PS1 mice were administered Ψ-GSH for 2 months starting at 8 month or 12 months of age. We show that Ψ-GSH treatment reduces indices of oxidative stress in older mice by restoration of enzyme glyoxalase-1 (Glo-1) activity and reduces levels of insoluble Aβ. Quantitative neuropathological analyses show that Ψ-GSH treatment significantly reduces Aβ deposition and brain inflammation in APP/PS1 mice compared to vehicle-treated mice. More importantly, Ψ-GSH treatment attenuated the progressive loss of cortical TH+ afferents and the loss of TH+ neurons in the locus coeruleus (LC). Collectively, the results show that Ψ-GSH exhibits significant antioxidant activity in aged APP/PS1 mice and chronic Ψ-GSH treatment administered after the onset of AD pathology can reverse/slow further progression of AD-like pathology and neurodegeneration in vivo.
Collapse
Affiliation(s)
- Ye In Christopher Kwon
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (Y.I.C.K.); (K.S.); (N.J.)
| | - Wei Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (W.X.); (H.Z.); (J.X.); (R.V.)
| | - Haizhou Zhu
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (W.X.); (H.Z.); (J.X.); (R.V.)
| | - Jiashu Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (W.X.); (H.Z.); (J.X.); (R.V.)
| | - Keaton Shinn
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (Y.I.C.K.); (K.S.); (N.J.)
| | - Nicholas Juckel
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (Y.I.C.K.); (K.S.); (N.J.)
| | - Robert Vince
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (W.X.); (H.Z.); (J.X.); (R.V.)
| | - Swati S. More
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (W.X.); (H.Z.); (J.X.); (R.V.)
- Correspondence: (S.S.M.); (M.K.L.)
| | - Michael K. Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (Y.I.C.K.); (K.S.); (N.J.)
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Correspondence: (S.S.M.); (M.K.L.)
| |
Collapse
|
47
|
Profiling the Concentration of Reduced and Oxidized Glutathione in Rat Brain Using HPLC/DAD Chromatographic System. Molecules 2021; 26:molecules26216590. [PMID: 34770998 PMCID: PMC8588517 DOI: 10.3390/molecules26216590] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/24/2021] [Accepted: 10/29/2021] [Indexed: 11/26/2022] Open
Abstract
This study aimed to develop a HPLC/DAD method in order to determine and quantify the reduced glutathione (GSH) and oxidized glutathione (GSSG) levels in rat brain. Due to the presence of the thiol group (-SH), GSH can interact with the Ellman′s reagent (DTNB), with which it forms a reaction product through which the level of GSH can be quantified, using the DAD detection system. Chromatographic separation was achieved after a derivatization process by using a mobile phase acetonitrile (A) and phosphate buffer (20 mM, pH = 2.5) (B). The compounds of interest were detected at 330 nm using a chromatographic C8 column. The method of determination met the validation criteria, specified by the regulatory bodies. The applicability of the method was demonstrated in a chronic toxicology study of central nervous system (CNS), following different treatment regimens with haloperidol.
Collapse
|
48
|
Anwar MM. Oxidative stress-A direct bridge to central nervous system homeostatic dysfunction and Alzheimer's disease. Cell Biochem Funct 2021; 40:17-27. [PMID: 34716723 DOI: 10.1002/cbf.3673] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/26/2022]
Abstract
Neurologists have highly observed a frequent increasing number of elderly patients with Alzheimer's disease (AD) without any relevant evidence of any genetic or known AD-linked predisposing factors in the past few years. Those patients are characterized by continuous and irreversible neuron cells loss along with declined cognitive functions. Numerous studies have suggested that the exaggerated release of reactive oxygen species (ROS) within the brain may develop late-onset neurodegenerative disorders, especially AD-neuroinflammatory type. However, the central nervous system is vitally linked with whole-brain chemical integrity and its related healthy state, the cascade by which ROS may result in AD's development has not been highly justified or even maintained. It is widely known that the brain consumes a vast amount of oxygen and is characterized by being rich in lipid polyunsaturated fatty acids content, explaining why it is a prone region to oxidative stress (OS) and ROS damage. The formed OS-AD cytoskeletal protein aggregates can be considered a main predisposing factor for amyloid-beta (Aβ) hallmarks precipitation. Herein, this review aims to provide a detailed information on how oxidative stress can play a pathogenic role in activating damage-associated molecular patterns (DAMPs)-related toll-like receptor-4 inflammatory (TLR-4) cascades resulting in the deposition of Aβ hallmarks in brain tissues ending with irreversible cognitive dysfunction. It also explains how microglia can be activated via ROS, which may significantly release several pro-inflammatory cascades ending with general brain atrophy. Furthermore, different types of suggested antioxidant therapies will be discussed to combat AD-related pathological disorders and hallmarks.
Collapse
Affiliation(s)
- Mai M Anwar
- Department of Biochemistry, National Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority (EDA), Cairo, Egypt.,Neuroscience Research Lab, Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| |
Collapse
|
49
|
Semenovich DS, Plotnikov EY, Titko OV, Lukiyenko EP, Kanunnikova NP. Effects of Panthenol and N-Acetylcysteine on Changes in the Redox State of Brain Mitochondria under Oxidative Stress In Vitro. Antioxidants (Basel) 2021; 10:antiox10111699. [PMID: 34829571 PMCID: PMC8614675 DOI: 10.3390/antiox10111699] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/16/2021] [Accepted: 10/24/2021] [Indexed: 11/16/2022] Open
Abstract
The glutathione system in the mitochondria of the brain plays an important role in maintaining the redox balance and thiol–disulfide homeostasis, whose violations are the important component of the biochemical shifts in neurodegenerative diseases. Mitochondrial dysfunction is known to be accompanied by the activation of free radical processes, changes in energy metabolism, and is involved in the induction of apoptotic signals. The formation of disulfide bonds is a leading factor in the folding and maintenance of the three-dimensional conformation of many specific proteins that selectively accumulate in brain structures during neurodegenerative pathology. In this study, we estimated brain mitochondria redox status and functioning during induction of oxidative damage in vitro. We have shown that the development of oxidative stress in vitro is accompanied by inhibition of energy metabolism in the brain mitochondria, a shift in the redox potential of the glutathione system to the oxidized side, and activation of S-glutathionylation of proteins. Moreover, we studied the effects of pantothenic acid derivatives—precursors of coenzyme A (CoA), primarily D-panthenol, that exhibit high neuroprotective activity in experimental models of neurodegeneration. Panthenol contributes to the significant restoration of the activity of enzymes of mitochondrial energy metabolism, normalization of the redox potential of the glutathione system, and a decrease in the level of S-glutathionylated proteins in brain mitochondria. The addition of succinate and glutathione precursor N-acetylcysteine enhances the protective effects of the drug.
Collapse
Affiliation(s)
- Dmitry S. Semenovich
- Institute of Biochemistry of Biologically Active Substances, NAS of Belarus, 230030 Grodno, Belarus; (O.V.T.); (E.P.L.); (N.P.K.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia;
- Correspondence: ; Tel.: +7-(925)-465-78-52
| | - Egor Yu. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia;
| | - Oksana V. Titko
- Institute of Biochemistry of Biologically Active Substances, NAS of Belarus, 230030 Grodno, Belarus; (O.V.T.); (E.P.L.); (N.P.K.)
| | - Elena P. Lukiyenko
- Institute of Biochemistry of Biologically Active Substances, NAS of Belarus, 230030 Grodno, Belarus; (O.V.T.); (E.P.L.); (N.P.K.)
| | - Nina P. Kanunnikova
- Institute of Biochemistry of Biologically Active Substances, NAS of Belarus, 230030 Grodno, Belarus; (O.V.T.); (E.P.L.); (N.P.K.)
- Department of Technology, Physiology and Food Hygiene, State University of Grodno, 230030 Grodno, Belarus
| |
Collapse
|
50
|
1,5-Benzodiazepin-2(3H)-ones: In Vitro Evaluation as Antiparkinsonian Agents. Antioxidants (Basel) 2021; 10:antiox10101584. [PMID: 34679721 PMCID: PMC8533176 DOI: 10.3390/antiox10101584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 01/08/2023] Open
Abstract
A new series of twenty-three 1,5-benzodiazepin-2(3H)-ones were synthesized and evaluated in the 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid (ABTS), ferric reducing antioxidant power (FRAP), and 2,2-diphenyl-1-picrylhydrazyl (DPPH) assays as a new chemotype with antioxidant and good drug-like properties. All of the derivatives showed low cytotoxicity in comparison to curcumin against the human neuroblastoma SH-SY5Y and the human hepatoma HepG2 cell lines. Experimental solubility in bio-relevant media showed a good relationship with melting points in this series. Five compounds with the best antioxidant properties showed neuroprotectant activity against H2O2-induced oxidative stress in the SH-SY5Y cell line. From them, derivatives 4-phenyl-1H-1,5-benzodiazepin-2(3H)-one (18) and 4-(3,4,5-trimethoxyphenyl)-1H-1,5-benzodiazepin-2(3H)-one (20) yielded good neuroprotection activity in the same neuronal cell line under 6-OHD and MPP+ insults as in vitro models of mitochondrial dysfunction and oxidative stress in Parkinson’s disease (PD). Both compounds also demonstrated a significant reduction of intracellular Reactive Oxygen Species (ROS) and superoxide levels, in parallel with a good improvement of the Mitochondrial Membrane Potential (ΔΨm). Compared with curcumin, compound 18 better reduced lipid peroxidation levels, malondialdehyde (MDA), in SH-SY5Y cells under oxidative stress pressure and recovered intracellular glutathione synthetase (GSH) levels. Apoptosis and caspase-3 levels of SH-SY5Y under H2O2 pressure were also reduced after treatment with 18. Neuroprotection in neuron-like differentiated SH-SY5Y cells was also achieved with 18. In summary, this family of 1,5-benzodiazepin-2-ones with an interesting antioxidant and drug-like profile, with low cytotoxic and good neuroprotectant activity, constitutes a new promising chemical class with high potential for the development of new therapeutic agents against PD.
Collapse
|