1
|
Gotoh S, Kawabori M, Yamaguchi S, Nakahara Y, Yoshie E, Konno K, Mizuno Y, Fujioka Y, Ohba Y, Kuge Y, Watanabe M, Fujimura M. Intranasal administration of stem cell-derived exosome alleviates cognitive impairment against subarachnoid hemorrhage. Exp Neurol 2025; 386:115143. [PMID: 39800250 DOI: 10.1016/j.expneurol.2025.115143] [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: 08/21/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
INTRODUCTION Brain damage caused by subarachnoid hemorrhage (SAH) currently lacks effective treatment, leading to stagnation in the improvement of functional outcomes for decades. Recent studies have demonstrated the therapeutic potential of exosomes released from mesenchymal stem cells (MSC), which effectively attenuate neuronal apoptosis and inflammation in neurological diseases. Due to the challenge of systemic dilution associated with intravenous administration, intranasal delivery has emerged as a novel approach for targeting the brain. In this study, we investigate the effects of intranasally administered MSC-derived exosomes in a SAH animal model and elucidate their mode of action. METHODS Exosomes were isolated from the cell supernatants of amnion-derived MSC. SAH was induced in 8-week-old Sprague-Dawley rats using an autologous blood prechiasmatic cistern injection model. A total of 1.2 × 1010 particles of exosomes in 200 μL of PBS or PBS alone were intranasally administered immediately and 24 h post-injury. Neurological function was assessed up to 7 days after injury, and histological analysis was performed to evaluate their anti-apoptotic and anti-inflammatory effects. The biodistribution of exosomes was assessed using PET/CT imaging of 64Cu labeled exosome. In vitro analyses were performed using primary glial cells and cell lines to evaluate the anti-inflammatory effects of the exosomes. RESULTS Animals treated with exosomes exhibited significant improvement in cognitive function compared with PBS treated animal. Apoptotic cells and inflammation were reduced for the exosome group in the hippocampal CA1 area and in cortex, resulting in better neuronal cell survival. Blood brain barrier permeability was also preserved in the exosome group. Nuclear imaging revealed that exosomes were primarily transferred to the olfactory nerve and cerebrum; furthermore, exosomes were also observed in the trigeminal nerve and brainstem, where exosomes were co-localized with microglia and with endothelial cells. In vitro assessment showed that exosome administration ameliorated inflammation and prevented the death of glial cells. CONCLUSIONS MSC-derived exosomes were successfully transferred into the brain through intranasal administration and alleviated brain damage following SAH.
Collapse
Affiliation(s)
- Shuho Gotoh
- Department of Neurosurgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Masahito Kawabori
- Department of Neurosurgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan.
| | - Sho Yamaguchi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Hyogo, Japan
| | - Yo Nakahara
- Department of Neurosurgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Erika Yoshie
- Department of Neurosurgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Kohtarou Konno
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yuki Mizuno
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan
| | - Yoichiro Fujioka
- Department of Cell Physiology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yusuke Ohba
- Department of Cell Physiology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Miki Fujimura
- Department of Neurosurgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
2
|
Shateeva VS, Simonenko SD, Khlystova MA, Selivanova EK, Borzykh AA, Gaynullina DK, Shvetsova AA. Perinatal hypoxia augments contractile impact of NADPH oxidase-derived ROS in early postnatal rat arteries. Pediatr Res 2025; 97:1220-1226. [PMID: 39127838 DOI: 10.1038/s41390-024-03466-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Reactive oxygen species (ROS), including those produced by NADPH oxidase (NOX), play an important vasomotor role, especially at early postnatal period. Mechanisms for regulating vascular tone can change significantly due to neonatal asphyxia and accompanying hypoxia. We tested the hypothesis that normobaric hypoxia (8% O2) for 2 h at the second day of life changes the functional contribution of NOX-derived ROS to the regulation of agonist-induced contraction in early postnatal rats. METHODS We studied saphenous arteries from 11- to 14-day-old male offspring using isometric myography and Western blotting and assessed the content of biochemical parameters in blood serum. RESULTS The values of main biochemical parameters in blood serum and the protein content of NOXs and superoxide dismutases in arterial tissue did not differ between "Control" and "Hypoxia" pups. The NOX inhibitor VAS2870 equally reduced the contractile responses of arteries to α1-adrenoceptor agonist methoxamine in "Control" and "Hypoxia" pups, but its effect was more pronounced in the arteries from "Hypoxia" pups when vasocontraction was evoked by the agonist of thromboxane A2 receptors U46619. CONCLUSION Perinatal hypoxia at the second day of life increases procontractile influence of NOX-derived ROS to the regulation of U46619-induced vasocontraction in the systemic arteries at early postnatal ontogenesis. IMPACT Nothing is known about programming effects of perinatal hypoxia, including birth asphyxia, on the ROS-mediated regulation of contraction in systemic arteries of early postnatal organism. 2-h normobaric hypoxia (8% O2) in rats at the second day of life increases the procontractile contribution of NOX-produced ROS to the regulation of U46619-induced vasocontraction in the systemic arteries at early postnatal ontogenesis. This fact may serve as a risk factor for the development of various disorders at later developmental stages and should be considered regarding the therapy for newborns who have suffered neonatal asphyxia.
Collapse
Affiliation(s)
- Valentina S Shateeva
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Sofia D Simonenko
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Margarita A Khlystova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Ekaterina K Selivanova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
- ChemRar Research and Development Institute, Khimki, Russia
| | - Anna A Borzykh
- Laboratory of Exercise Physiology, State Research Center of the Russian Federation-Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Dina K Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Anastasia A Shvetsova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
3
|
Yang Y, Chen S, Zhang L, Zhang G, Liu Y, Li Y, Zou L, Meng L, Tian Y, Dai L, Xiong M, Pan L, Xiong J, Chen L, Hou H, Yu Z, Zhang Z. The PM20D1-NADA pathway protects against Parkinson's disease. Cell Death Differ 2024; 31:1545-1560. [PMID: 39174646 PMCID: PMC11519464 DOI: 10.1038/s41418-024-01356-9] [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: 05/25/2023] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024] Open
Abstract
Parkinson's disease (PD) is characterized by the selective loss of dopaminergic neurons in the substantia nigra and the accumulation of α-synuclein (α-Syn) aggregates. However, the molecular mechanisms regulating α-Syn aggregation and neuronal degeneration remain poorly understood. The peptidase M20 domain containing 1 (PM20D1) gene lies within the PARK16 locus genetically linked to PD. Single nucleotide polymorphisms regulating PM20D1 expression are associated with changed risk of PD. Dopamine (DA) metabolism and DA metabolites have been reported to regulate α-Syn pathology. Here we report that PM20D1 catalyzes the conversion of DA to N-arachidonoyl dopamine (NADA), which interacts with α-Syn and inhibits its aggregation. Simultaneously, NADA competes with α-Syn fibrils to regulate TRPV4-mediated calcium influx and downstream phosphatases, thus alleviating α-Syn phosphorylation. The expression of PM20D1 decreases during aging. Overexpression of PM20D1 or the administration of NADA in a mouse model of synucleinopathy alleviated α-Syn pathology, dopaminergic neurodegeneration, and motor impairments. These observations support the protective effect of the PM20D1-NADA pathway against the progression of α-Syn pathology in PD.
Collapse
Affiliation(s)
- Yunying Yang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Sichun Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Li Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guoxin Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yan Liu
- Department of Nursing, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiming Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Li Zou
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Tian
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Min Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lina Pan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liam Chen
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Hua Hou
- Department of Polymer Science, College of Chemistry and Molecular Sciences of Wuhan University, Wuhan, 430060, China
| | - Zhui Yu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430000, China.
| |
Collapse
|
4
|
Huo L, Fu J, Wang S, Wang H, Liu X. Emerging ferroptosis inhibitors as a novel therapeutic strategy for the treatment of neonatal hypoxic-ischemic encephalopathy. Eur J Med Chem 2024; 271:116453. [PMID: 38701713 DOI: 10.1016/j.ejmech.2024.116453] [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: 08/01/2023] [Revised: 08/30/2023] [Accepted: 04/24/2024] [Indexed: 05/05/2024]
Abstract
Neonatal hypoxia-ischemia encephalopathy (NHIE), an oxygen deprivation-mediated brain injury due to birth asphyxia or reduced cerebral blood perfusion, often leads to lifelong sequelae, including seizures, cerebral palsy, and mental retardation. NHIE poses a significant health challenge, as one of the leading causes of neonatal morbidity and mortality globally. Despite this, available therapies are limited. Numerous studies have recently demonstrated that ferroptosis, an iron-dependent non-apoptotic regulated form of cell death characterized by lipid peroxidation (LPO) and iron dyshomeostasis, plays a role in the genesis of NHIE. Moreover, recently discovered compounds have been shown to exert potential therapeutic effects on NHIE by inhibiting ferroptosis. This comprehensive review summarizes the fundamental mechanisms of ferroptosis contributing to NHIE. We focus on various emerging therapeutic compounds exhibiting characteristics of ferroptosis inhibition and delineate their pharmacological benefits for the treatment of NHIE. This review suggests that pharmacological inhibition of ferroptosis may be a potential therapeutic strategy for NHIE.
Collapse
Affiliation(s)
- Liang Huo
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 11004, China.
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 11004, China
| | - Shimeng Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 11004, China
| | - Hua Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 11004, China
| | - Xueyan Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 11004, China.
| |
Collapse
|
5
|
Gedzun VR, Khukhareva DD, Sarycheva NY, Kotova MM, Kabiolsky IA, Dubynin VA. Perinatal Stressors as a Factor in Impairments to Nervous System Development and Functions: Review of In Vivo Models. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2023; 53:61-69. [PMID: 36969360 PMCID: PMC10006566 DOI: 10.1007/s11055-023-01391-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 04/26/2022] [Indexed: 03/24/2023]
Abstract
The human body is faced with stress throughout ontogeny. At the stage of intrauterine development, the mother’s body serves as a source of resources and most of the humoral factors supporting the development of the fetus. In normal conditions, maternal stress-related humoral signals (e.g., cortisol) regulate fetal development; however, distress (excessive pathological stress) in the perinatal period leads to serious and sometimes irreversible changes in the developing brain. The mother being in an unfavorable psychoemotional state, toxins and teratogens, environmental conditions, and severe infectious diseases are the most common risk factors for the development of perinatal nervous system pathology in the modern world. In this regard, the challenge of modeling situations in which prenatal or early postnatal stresses lead to serious impairments to brain development and functioning is extremely relevant. This review addresses the various models of perinatal pathology used in our studies (hypoxia, exposure to valproate, hyperserotoninemia, alcoholization), and assesses the commonality of the mechanisms of the resulting disorders and behavioral phenotypes forming in these models, as well as their relationship with models of perinatal pathology based on the impact of psychoemotional stressors.
Collapse
Affiliation(s)
- V. R. Gedzun
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - D. D. Khukhareva
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - N. Yu. Sarycheva
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - M. M. Kotova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - I. A. Kabiolsky
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - V. A. Dubynin
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
6
|
Bandala C, Cárdenas-Rodríguez N, Mendoza-Torreblanca JG, Contreras-García IJ, Martínez-López V, Cruz-Hernández TR, Carro-Rodríguez J, Vargas-Hernández MA, Ignacio-Mejía I, Alfaro-Rodriguez A, Lara-Padilla E. Therapeutic Potential of Dopamine and Related Drugs as Anti-Inflammatories and Antioxidants in Neuronal and Non-Neuronal Pathologies. Pharmaceutics 2023; 15:pharmaceutics15020693. [PMID: 36840015 PMCID: PMC9966027 DOI: 10.3390/pharmaceutics15020693] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/11/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Dopamine (DA), its derivatives, and dopaminergic drugs are compounds widely used in the management of diseases related to the nervous system. However, DA receptors have been identified in nonneuronal tissues, which has been related to their therapeutic potential in pathologies such as sepsis or septic shock, blood pressure, renal failure, diabetes, and obesity, among others. In addition, DA and dopaminergic drugs have shown anti-inflammatory and antioxidant properties in different kinds of cells. AIM To compile the mechanism of action of DA and the main dopaminergic drugs and show the findings that support the therapeutic potential of these molecules for the treatment of neurological and non-neurological diseases considering their antioxidant and anti-inflammatory actions. METHOD We performed a review article. An exhaustive search for information was carried out in specialized databases such as PubMed, PubChem, ProQuest, EBSCO, Scopus, Science Direct, Web of Science, Bookshelf, DrugBank, Livertox, and Clinical Trials. RESULTS We showed that DA and dopaminergic drugs have emerged for the management of neuronal and nonneuronal diseases with important therapeutic potential as anti-inflammatories and antioxidants. CONCLUSIONS DA and DA derivatives can be an attractive treatment strategy and a promising approach to slowing the progression of disorders through repositioning.
Collapse
Affiliation(s)
- Cindy Bandala
- Neurociencia Básica, Instituto Nacional de Rehabilitación LGII, Secretaría de Salud, Mexico City 14389, Mexico
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Correspondence: (C.B.); (E.L.-P.); Tel.: +52-(55)-5999-1000 (ext. 19307) (C.B.); +52-(55)-57296000 (ext. 62712) (E.L.-P.)
| | - Noemi Cárdenas-Rodríguez
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Mexico City 04530, Mexico
| | | | | | - Valentín Martínez-López
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico
| | | | - Jazmín Carro-Rodríguez
- Escuela de Biología Experimental, Unidad Iztapalapa, Universidad Autónoma Metropolitana, Mexico City 09340, Mexico
| | | | - Iván Ignacio-Mejía
- Laboratorio de Medicina Traslacional, Escuela Militar de Graduados de Sanidad, Mexico City 11200, Mexico
| | - Alfonso Alfaro-Rodriguez
- Neurociencia Básica, Instituto Nacional de Rehabilitación LGII, Secretaría de Salud, Mexico City 14389, Mexico
| | - Eleazar Lara-Padilla
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Correspondence: (C.B.); (E.L.-P.); Tel.: +52-(55)-5999-1000 (ext. 19307) (C.B.); +52-(55)-57296000 (ext. 62712) (E.L.-P.)
| |
Collapse
|
7
|
Influence of N-Arachidonoyl Dopamine and N-Docosahexaenoyl Dopamine on the Expression of Neurotrophic Factors in Neuronal Differentiated Cultures of Human Induced Pluripotent Stem Cells under Conditions of Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11010142. [PMID: 35052646 PMCID: PMC8773408 DOI: 10.3390/antiox11010142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 01/27/2023] Open
Abstract
Oxidative stress (OS) is implicated in the pathogenesis of several neurodegenerative diseases. We have previously shown that N-acyl dopamines (N-ADA and N-DDA) protect the neural cells of healthy donors and patients with Parkinson’s disease from OS. In this study, we assessed the effects of N-acyl dopamines on the expression of neurotrophic factors in human-induced pluripotent stem cell-derived neuronal cultures enriched with dopaminergic neurons under conditions of OS induced by hydrogen peroxide. We showed that hydrogen peroxide treatment increased BDNF but not GDNF mRNA levels, while it did not affect the secretion of corresponding proteins into the culture medium of these cells. Application of N-acyl dopamines promoted BDNF release into the culture medium. Under conditions of OS, N-DDA also increased TRKB, TRKC and RET mRNA levels. Furthermore, N-acyl dopamines prevented cell death 24 h after OS induction and promoted the expression of antioxidant enzymes GPX1, GPX7, SOD1, SOD2 and CAT, as well as reduced the BAX/BCL2 mRNA ratio. These findings indicate that stimulation of the expression of neurotrophic factors and their receptors may underlie the neuroprotective effects of N-acyl dopamines in human neurons.
Collapse
|
8
|
Ladak Z, Garcia E, Yoon J, Landry T, Armstrong EA, Yager JY, Persad S. Sulforaphane (SFA) protects neuronal cells from oxygen & glucose deprivation (OGD). PLoS One 2021; 16:e0248777. [PMID: 33735260 PMCID: PMC7971874 DOI: 10.1371/journal.pone.0248777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 03/04/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Perinatal brain injury results in neurodevelopmental disabilities (neuroDDs) that include cerebral palsy, autism, attention deficit disorder, epilepsy, learning disabilities and others. Commonly, injury occurs when placental circulation, that is responsible for transporting nutrients and oxygen to the fetus, is compromised. Placental insufficiency (PI) is a reduced supply of blood and oxygen to the fetus and results in a hypoxic-ischemic (HI) environment. A significant HI state in-utero leads to perinatal compromise, characterized by fetal growth restriction and brain injury. Given that over 80% of perinatal brain injuries that result in neuroDDs occur during gestation, prior to birth, preventive approaches are needed to reduce or eliminate the potential for injury and subsequent neuroDDs. Sulforaphane (SFA) derived from cruciferous vegetables such as broccoli sprouts (BrSps) is a phase-II enzyme inducer that acts via cytoplasmic Nrf2 to enhance the production of anti-oxidants in the brain through the glutathione pathway. We have previously shown a profound in vivo neuro-protective effect of BrSps/SFA as a dietary supplement in pregnant rat models of both PI and fetal inflammation. Strong evidence also points to a role for SFA as treatment for various cancers. Paradoxically, then SFA has the ability to enhance cell survival, and with conditions of cancer, enhance cell death. Given our findings of the benefit of SFA/Broccoli Sprouts as a dietary supplement during pregnancy, with improvement to the fetus, it is important to determine the beneficial and toxic dosing range of SFA. We therefore explored, in vitro, the dosing range of SFA for neuronal and glial protection and toxicity in normal and oxygen/glucose deprived (OGD) cell cultures. METHODS OGD simulates, in vitro, the condition experienced by the fetal brain due to PI. We developed a cell culture model of primary cortical neuronal, astrocyte and combined brain cell co-cultures from newborn rodent brains. The cultures were exposed to an OGD environment for various durations of time to determine the LD50 (duration of OGD required for 50% cell death). Using the LD50 as the time point, we evaluated the efficacy of varying doses of SFA for neuroprotective and neurotoxicity effects. Control cultures were exposed to normal media without OGD, and cytotoxicity of varying doses of SFA was also evaluated. Immunofluorescence (IF) and Western blot analysis of cell specific markers were used for culture characterization, and quantification of LD50. Efficacy and toxicity effect of SFA was assessed by IF/high content microscopy and by AlamarBlue viability assay, respectively. RESULTS We determined the LD50 to be 2 hours for neurons, 8 hours for astrocytes, and 10 hours for co-cultures. The protective effect of SFA was noticeable at 2.5 μM and 5 μM for neurons, although it was not significant. There was a significant protective effect of SFA at 2.5 μM (p<0.05) for astrocytes and co-cultures. Significant toxicity ranges were also confirmed in OGD cultures as ≥ 100 μM (p<0.05) for astrocytes, ≥ 50 μM (p<0.01) for co-cultures, but not toxic in neurons; and toxic in control cultures as ≥ 100 μM (p<0.01) for neurons, and ≥ 50 μM (p<0.01) for astrocytes and co-cultures. One Way ANOVA and Dunnett's Multiple Comparison Test were used for statistical analysis. CONCLUSIONS Our results indicate that cell death shows a trend to reduction in neuronal and astrocyte cultures, and is significantly reduced in co-cultures treated with low doses of SFA exposed to OGD. Doses of SFA that were 10 times higher were toxic, not only under conditions of OGD, but in normal control cultures as well. The findings suggest that: 1. SFA shows promise as a preventative agent for fetal ischemic brain injury, and 2. Because the fetus is a rapidly growing organism with profound cell multiplication, dosing parameters must be established to insure safety within efficacious ranges. This study will influence the development of innovative therapies for the prevention of childhood neuroDD.
Collapse
Affiliation(s)
- Zeenat Ladak
- Faculty of Medicine & Dentistry, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Elizabeth Garcia
- Faculty of Medicine & Dentistry, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Jenny Yoon
- Faculty of Medicine & Dentistry, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Takaaki Landry
- Faculty of Medicine & Dentistry, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Edward A. Armstrong
- Faculty of Medicine & Dentistry, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Jerome Y. Yager
- Faculty of Medicine & Dentistry, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Sujata Persad
- Faculty of Medicine & Dentistry, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
9
|
Dobrovolsky AP, Gedzun VR, Bogin VI, Ma D, Ichim TE, Sukhanova IA, Malyshev AV, Dubynin VA. Beneficial effects of xenon inhalation on behavioral changes in a valproic acid-induced model of autism in rats. J Transl Med 2019; 17:400. [PMID: 31796043 PMCID: PMC6891980 DOI: 10.1186/s12967-019-02161-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 11/27/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Xenon (Xe) is a noble gas that has been used for the last several decades as an anesthetic during surgery. Its antagonistic effect on glutamate subtype of NMDA (N-methyl-D-aspartate) receptors resulted in evaluation of this gas for treatment of CNS pathologies, including psychoemotional disorders. The aim of this study was to assess the behavioral effects of acute inhalation of subanesthetic concentrations of Xe and to study the outcomes of Xe exposure in valproic acid (VPA)-induced rodent model of autism. METHODS We have conducted two series of experiments with a battery of behavioral tests aimed to evaluate locomotion, anxiety- and depression-like behavior, and social behavior in healthy, VPA-treated and Xe-exposed young rats. RESULTS We have shown that in healthy animals Xe exposure resulted in acute and delayed decrease of exploratory motivation, partial decrease in risk-taking and depressive-like behavior as well as improved sensorimotor integration during the negative geotaxis test. Acute inhalations of Xe in VPA-exposed animals led to improvement in social behavior, decrease in exploratory motivation, and normalization of behavior in forced-swim test. CONCLUSION Behavioral modulatory effects of Xe are probably related to its generalized action on excitatory/inhibitory balance within the CNS. Our data suggest that subanesthetic short-term exposures to Xe have beneficial effect on several behavioral modalities and deserves further investigation.
Collapse
Affiliation(s)
- A P Dobrovolsky
- Pirogov Russian National Research Medical University, Ostrovitianov str. 1, Moscow, 117997, Russia.
| | - V R Gedzun
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - V I Bogin
- Nobilis Therapeutics Inc, Portland, OR, USA
| | - D Ma
- Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery & Cancer, Imperial College London, London, UK
| | - T E Ichim
- Nobilis Therapeutics Inc, Portland, OR, USA
| | - Iu A Sukhanova
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - A V Malyshev
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - V A Dubynin
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|