1
|
Herpich ME, de Oliveira Guarnieri L, de Oliveira ACP, Moraes MFD. Bacterial Lipopolysaccharide Post-Conditioning in The kainic acid animal model of Temporal Lobe epilepsy. Epilepsy Behav 2024; 161:110076. [PMID: 39467457 DOI: 10.1016/j.yebeh.2024.110076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024]
Abstract
This study used intra-hippocampal injections of Kainic Acid (KA) in Wistar rats to induce spontaneous recurrent seizures (SRS) after a 9-day latent period. A post-conditioning protocol with LPS, injected at the same site 72 h after the initial KA insult, was employed to trigger secondary competing processes. To evaluate the post-conditioning effect of LPS, 25 animals were divided into four groups: SAL-SAL (n = 6), KA-SAL (n = 6), SAL-LPS (n = 7), and KA-LPS (n = 6). SRS occurrence and seizure duration were quantified through video monitoring from days 9 to 17, along with other ictal behaviors, such as tail-chasing and wet-dog-shakes. Behavioral assessments revealed that the KA-LPS group had preserved sucrose preference and intact long-term memory in the object recognition test, indicating reduced depressive-like behavior and cognitive preservation compared to the KA-SAL group. The forced swim test showed increased depressive-like behavior in the SAL-LPS group, with LPS mitigating these effects in the KA group. The marble-burying test showed no significant differences among groups. Animals were euthanized on day 26, and hippocampal slices were analyzed using fluoro-jade staining for cell death and immunofluorescence staining for Iba-1 (microglia) and GFAP (astrocyte) labeling. The results support the hypothesis that epileptogenesis involves a cascade of plastic changes in neural networks and that precise, timely interventions can potentially interfere with this process.
Collapse
Affiliation(s)
- Mateus Eduardo Herpich
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo de Oliveira Guarnieri
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Centro de Tecnologia e Pesquisa em Magneto Ressonância, Programa de Pós-Graduação em Engenharia Elétrica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Márcio Flávio Dutra Moraes
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Centro de Tecnologia e Pesquisa em Magneto Ressonância, Programa de Pós-Graduação em Engenharia Elétrica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
2
|
Li E, Yan R, Qiao H, Sun J, Zou P, Chang J, Li S, Ma Q, Zhang R, Liao B. Combined transcriptomics and proteomics studies on the effect of electrical stimulation on spinal cord injury in rats. Heliyon 2024; 10:e23960. [PMID: 38226269 PMCID: PMC10788535 DOI: 10.1016/j.heliyon.2023.e23960] [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: 03/02/2023] [Revised: 11/20/2023] [Accepted: 12/19/2023] [Indexed: 01/17/2024] Open
Abstract
Electrical stimulation (ES) of the spinal cord is a promising therapy for functional rehabilitation after spinal cord injury (SCI). However, the specific mechanism of action is poorly understood. We designed and applied an implanted ES device in the SCI area in rats and determined the effect of ES on the treatment of motor dysfunction after SCI using behavioral scores. Additionally, we examined the molecular characteristics of the samples using proteomic and transcriptomic sequencing. The differential molecules between groups were identified using statistical analyses. Molecular, network, and pathway-based analyses were used to identify group-specific biological features. ES (0.5 mA, 0.1 ms, 50 Hz) had a positive effect on motor dysfunction and neuronal regeneration in rats after SCI. Six samples (three independent replicates in each group) were used for transcriptome sequencing; we obtained 1026 differential genes, comprising 274 upregulated genes and 752 downregulated genes. A total of 10 samples were obtained: four samples in the ES group and six samples in the SCI group; for the proteome sequencing, 48 differential proteins were identified, including 45 up-regulated and three down-regulated proteins. Combined transcriptomic and proteomic studies have shown that the main enrichment pathway is the hedgehog signaling pathway. Western blot results showed that the expression levels of Sonic hedgehog (SHH) (P < 0.001), Smoothened (SMO) (P = 0.0338), and GLI-1 (P < 0.01) proteins in the ES treatment group were significantly higher than those in the SCI group. The immunofluorescence results showed significantly increased expression of SHH (P = 0.0181), SMO (P = 0.021), and GLI-1 (P = 0.0126) in the ES group compared with that in the SCI group. In conclusion, ES after SCI had a positive effect on motor dysfunction and anti-inflammatory effects in rats. Moreover, transcriptomic and proteomic sequencing also provided unique perspectives on the complex relationships between ES on SCI, where the SHH signaling pathway plays a critical role. Our study provides a significant theoretical foundation for the clinical implementation of ES therapy in patients with SCI.
Collapse
Affiliation(s)
- Erliang Li
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Rongbao Yan
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huanhuan Qiao
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Jin Sun
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Peng Zou
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiaqi Chang
- School of Automation Science and Electrical Engineering, Beihang University, 37th Xueyuan Road, Beijing, China
| | - Shuang Li
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Qiong Ma
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Rui Zhang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Bo Liao
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
3
|
Kalinichenko SG, Pushchin II, Matveeva NY. Neurotoxic and cytoprotective mechanisms in the ischemic neocortex. J Chem Neuroanat 2023; 128:102230. [PMID: 36603664 DOI: 10.1016/j.jchemneu.2022.102230] [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: 09/19/2022] [Revised: 12/30/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Neuronal damage in ischemic stroke occurs due to permanent imbalance between the metabolic needs of the brain and the ability of the blood-vascular system to maintain glucose delivery and adequate gas exchange. Oxidative stress and excitotoxicity trigger complex processes of neuroinflammation, necrosis, and apoptosis of both neurons and glial cells. This review summarizes data on the structural and chemical changes in the neocortex and main cytoprotective effects induced by focal ischemic stroke. We focus on the expression of neurotrophins (NT) and molecular and cellular changes in neurovascular units in ischemic brain. We also discuss how these factors affect the apoptosis of cortical cells. Ischemic damage involves close interaction of a wide range of signaling molecules, each acting as an efficient marker of cell state in both the ischemic core and penumbra. NTs play the main regulatory role in brain tissue recovery after ischemic injury. Heterogeneous distribution of the BDNF, NT-3, and GDNF immunoreactivity is concordant with the selective response of different types of cortical neurons and glia to ischemic injury and allows mapping the position of viable neurons. Astrocytes are the central link in neurovascular coupling in ischemic brain by providing other cells with a wide range of vasotropic factors. The NT expression coincides with the distribution of reactive astrocytes, marking the boundaries of the penumbra. The development of ischemic stroke is accompanied by a dramatic change in the distribution of GDNF reactivity. In early ischemic period, it is mainly observed in cortical neurons, while in late one, the bulk of GDNF-positive cells are various types of glia, in particular, astrocytes. The proportion of GDNF-positive astrocytes increases gradually throughout the ischemic period. Some factors that exert cytoprotective effects in early ischemic period may display neurotoxic and pro-apoptotic effects later on. The number of apoptotic cells in the ischemic brain tissue correlates with the BDNF levels, corroborating its protective effects. Cytoprotection and neuroplasticity are two lines of brain protection and recovery after ischemic stroke. NTs can be considered an important link in these processes. To develop efficient pharmacological therapy for ischemic brain injury, we have to deepen our understanding of neurochemical adaptation of brain tissue to acute stroke.
Collapse
Affiliation(s)
- Sergei G Kalinichenko
- Department of Histology, Cytology, and Embryology, Pacific State Medical University, Vladivostok 690950, Russia
| | - Igor I Pushchin
- Laboratory of Physiology, A.V. Zhirmusky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia.
| | - Natalya Yu Matveeva
- Department of Histology, Cytology, and Embryology, Pacific State Medical University, Vladivostok 690950, Russia
| |
Collapse
|
4
|
Kim S, Pajarillo E, Nyarko-Danquah I, Aschner M, Lee E. Role of Astrocytes in Parkinson's Disease Associated with Genetic Mutations and Neurotoxicants. Cells 2023; 12:622. [PMID: 36831289 PMCID: PMC9953822 DOI: 10.3390/cells12040622] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/02/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons and the aggregation of Lewy bodies in the basal ganglia, resulting in movement impairment referred to as parkinsonism. However, the etiology of PD is not well known, with genetic factors accounting only for 10-15% of all PD cases. The pathogenetic mechanism of PD is not completely understood, although several mechanisms, such as oxidative stress and inflammation, have been suggested. Understanding the mechanisms of PD pathogenesis is critical for developing highly efficacious therapeutics. In the PD brain, dopaminergic neurons degenerate mainly in the basal ganglia, but recently emerging evidence has shown that astrocytes also significantly contribute to dopaminergic neuronal death. In this review, we discuss the role of astrocytes in PD pathogenesis due to mutations in α-synuclein (PARK1), DJ-1 (PARK7), parkin (PARK2), leucine-rich repeat kinase 2 (LRRK2, PARK8), and PTEN-induced kinase 1 (PINK1, PARK6). We also discuss PD experimental models using neurotoxins, such as paraquat, rotenone, 6-hydroxydopamine, and MPTP/MPP+. A more precise and comprehensive understanding of astrocytes' modulatory roles in dopaminergic neurodegeneration in PD will help develop novel strategies for effective PD therapeutics.
Collapse
Affiliation(s)
- Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
5
|
Zhu Z, Quadri Z, Crivelli SM, Elsherbini A, Zhang L, Tripathi P, Qin H, Roush E, Spassieva SD, Nikolova-Karakashian M, McClintock TS, Bieberich E. Neutral Sphingomyelinase 2 Mediates Oxidative Stress Effects on Astrocyte Senescence and Synaptic Plasticity Transcripts. Mol Neurobiol 2022; 59:3233-3253. [PMID: 35294731 PMCID: PMC9023069 DOI: 10.1007/s12035-022-02747-0] [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: 08/28/2021] [Accepted: 01/11/2022] [Indexed: 11/26/2022]
Abstract
We have shown that deficiency of neutral sphingomyelinase 2 (nSMase2), an enzyme generating the sphingolipid ceramide, improves memory in adult mice. Here, we performed sphingolipid and RNA-seq analyses on the cortex from 10-month-old nSMase2-deficient (fro/fro) and heterozygous (+ /fro) mice. fro/fro cortex showed reduced levels of ceramide, particularly in astrocytes. Differentially abundant transcripts included several functionally related groups, with decreases in mitochondrial oxidative phosphorylation and astrocyte activation transcripts, while axon guidance and synaptic transmission and plasticity transcripts were increased, indicating a role of nSMase2 in oxidative stress, astrocyte activation, and cognition. Experimentally induced oxidative stress decreased the level of glutathione (GSH), an endogenous inhibitor of nSMase2, and increased immunolabeling for ceramide in primary + /fro astrocytes, but not in fro/fro astrocytes. β-galactosidase activity was lower in 5-week-old fro/fro astrocytes, indicating delayed senescence due to nSMase2 deficiency. In fro/fro cortex, levels of the senescence markers C3b and p27 and the proinflammatory cytokines interleukin 1β, interleukin 6, and tumor necrosis factor α were reduced, concurrent with twofold decreased phosphorylation of their downstream target, protein kinase Stat3. RNA and protein levels of the ionotropic glutamate receptor subunit 2B (Grin2b/NR2B) were increased by twofold, which was previously shown to enhance cognition. This was consistent with threefold reduced levels of exosomes carrying miR-223-3p, a micro-RNA downregulating NR2B. In summary, our data show that nSMase2 deficiency prevents oxidative stress-induced elevation of ceramide and secretion of exosomes by astrocytes that suppress neuronal function, indicating a role of nSMase2 in the regulation of neuroinflammation and cognition.
Collapse
Affiliation(s)
- Zhihui Zhu
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Zainuddin Quadri
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
- Veterans Affairs Medical Center, Lexington, KY 40502, United States
| | - Simone M. Crivelli
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Ahmed Elsherbini
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Liping Zhang
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
- Veterans Affairs Medical Center, Lexington, KY 40502, United States
| | - Priyanka Tripathi
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
- Veterans Affairs Medical Center, Lexington, KY 40502, United States
| | - Haiyan Qin
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Emily Roush
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Stefka D. Spassieva
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | | | - Timothy S. McClintock
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Erhard Bieberich
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
- Veterans Affairs Medical Center, Lexington, KY 40502, United States
| |
Collapse
|
6
|
Albers JL, Steibel JP, Klingler RH, Ivan LN, Garcia-Reyero N, Carvan MJ, Murphy CA. Altered Larval Yellow Perch Swimming Behavior Due to Methylmercury and PCB126 Detected Using Hidden Markov Chain Models. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:3514-3523. [PMID: 35201763 DOI: 10.1021/acs.est.1c07505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Fish swimming behavior is a commonly measured response in aquatic ecotoxicology because behavior is considered a whole organism-level effect that integrates many sensory systems. Recent advancements in animal behavior models, such as hidden Markov chain models (HMM), suggest an improved analytical approach for toxicology. Using both new and traditional approaches, we examined the sublethal effects of PCB126 and methylmercury on yellow perch (YP) larvae (Perca flavescens) using three doses. Both approaches indicate larvae increase activity after exposure to either chemical. The middle methylmercury-dosed larvae showed multiple altered behavior patterns. First, larvae had a general increase in activity, typically performing more behavior states, more time swimming, and more swimming bouts per second. Second, when larvae were in a slow or medium swimming state, these larvae tended to switch between these states more often. Third, larvae swam slower during the swimming bouts. The upper PCB126-dosed larvae exhibited a higher proportion and a fast swimming state, but the total time spent swimming fast decreased. The middle PCB126-dosed larvae transitioned from fast to slow swimming states less often than the control larvae. These results indicate that developmental exposure to very low doses of these neurotoxicants alters YP larvae overall swimming behaviors, suggesting neurodevelopment alteration.
Collapse
Affiliation(s)
- Janice L Albers
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, Michigan 48824, United States
| | - Juan P Steibel
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, Michigan 48824, United States
| | - Rebekah H Klingler
- School of Freshwater Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53204, United States
| | - Lori N Ivan
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, Michigan 48824, United States
| | - Natàlia Garcia-Reyero
- Environmental Laboratory, US Army Engineer Research and Development Center, Vicksburg, Mississippi, 39180, United States
| | - Michael J Carvan
- School of Freshwater Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53204, United States
| | - Cheryl A Murphy
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
7
|
Glutamate Uptake Is Not Impaired by Hypoxia in a Culture Model of Human Fetal Neural Stem Cell-Derived Astrocytes. Genes (Basel) 2022; 13:genes13030506. [PMID: 35328060 PMCID: PMC8953426 DOI: 10.3390/genes13030506] [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: 01/30/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
Hypoxic ischemic injury to the fetal and neonatal brain is a leading cause of death and disability worldwide. Although animal and culture studies suggest that glutamate excitotoxicity is a primary contributor to neuronal death following hypoxia, the molecular mechanisms, and roles of various neural cells in the development of glutamate excitotoxicity in humans, is not fully understood. In this study, we developed a culture model of human fetal neural stem cell (FNSC)-derived astrocytes and examined their glutamate uptake in response to hypoxia. We isolated, established, and characterized cultures of FNSCs from aborted fetal brains and differentiated them into astrocytes, characterized by increased expression of the astrocyte markers glial fibrillary acidic protein (GFAP), excitatory amino acid transporter 1 (EAAT1) and EAAT2, and decreased expression of neural stem cell marker Nestin. Differentiated astrocytes were exposed to various oxygen concentrations mimicking normoxia (20% and 6%), moderate and severe hypoxia (2% and 0.2%, respectively). Interestingly, no change was observed in the expression of the glutamate transporter EAAT2 or glutamate uptake by astrocytes, even after exposure to severe hypoxia for 48 h. These results together suggest that human FNSC-derived astrocytes can maintain glutamate uptake after hypoxic injury and thus provide evidence for the possible neuroprotective role of astrocytes in hypoxic conditions.
Collapse
|
8
|
Jaganjac M, Milkovic L, Zarkovic N, Zarkovic K. Oxidative stress and regeneration. Free Radic Biol Med 2022; 181:154-165. [PMID: 35149216 DOI: 10.1016/j.freeradbiomed.2022.02.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/06/2022] [Indexed: 12/19/2022]
Abstract
Regeneration is the process of replacing/restoring a damaged cell/tissue/organ to its full function and is limited respecting complexity of specific organ structures and the level of differentiation of the cells. Unlike physiological cell turnover, this tissue replacement form is activated upon pathological stimuli such as injury and/or disease that usually involves inflammatory response. To which extent will tissue repair itself depends on many factors and involves different mechanisms. Oxidative stress is one of them, either acute, as in case of traumatic brin injury or chronic, as in case of neurodegeneration, oxidative stress within brain involves lipid peroxidation, which generates reactive aldehydes, such as 4-hydroxynonenal (4-HNE). While 4-HNE is certainly neurotoxic and causes disruption of the blood brain barrier in case of severe injuries, it is also physiologically produced by glial cells, especially astrocytes, but its physiological roles within CNS are not understood. Because 4-HNE can regulate the response of the other cells in the body to stress, enhance their antioxidant capacities, proliferation and differentiation, we could assume that it may also have some beneficial role for neuroregeneration. Therefore, future studies on the relevance of 4-HNE for the interaction between neuronal cells, notably stem cells and reactive astrocytes might reveal novel options to better monitor and treat consequences or brain injuries, neurodegeneration and regeneration.
Collapse
Affiliation(s)
- Morana Jaganjac
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress (LabOS), Div. Molecular Medicine, Bijenicka 54, Zagreb, Croatia
| | - Lidija Milkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress (LabOS), Div. Molecular Medicine, Bijenicka 54, Zagreb, Croatia
| | - Neven Zarkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress (LabOS), Div. Molecular Medicine, Bijenicka 54, Zagreb, Croatia.
| | - Kamelija Zarkovic
- University of Zagreb, School of Medicine, Div. of Pathology, Neuropathology Unit, University Hospital Centre Zagreb, Kispaticeva 12, Zagreb, Croatia
| |
Collapse
|
9
|
Quincozes-Santos A, Santos CL, de Souza Almeida RR, da Silva A, Thomaz NK, Costa NLF, Weber FB, Schmitz I, Medeiros LS, Medeiros L, Dotto BS, Dias FRP, Sovrani V, Bobermin LD. Gliotoxicity and Glioprotection: the Dual Role of Glial Cells. Mol Neurobiol 2021; 58:6577-6592. [PMID: 34581988 PMCID: PMC8477366 DOI: 10.1007/s12035-021-02574-9] [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: 07/22/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023]
Abstract
Glial cells (astrocytes, oligodendrocytes and microglia) are critical for the central nervous system (CNS) in both physiological and pathological conditions. With this in mind, several studies have indicated that glial cells play key roles in the development and progression of CNS diseases. In this sense, gliotoxicity can be referred as the cellular, molecular, and neurochemical changes that can mediate toxic effects or ultimately lead to impairment of the ability of glial cells to protect neurons and/or other glial cells. On the other hand, glioprotection is associated with specific responses of glial cells, by which they can protect themselves as well as neurons, resulting in an overall improvement of the CNS functioning. In addition, gliotoxic events, including metabolic stresses, inflammation, excitotoxicity, and oxidative stress, as well as their related mechanisms, are strongly associated with the pathogenesis of neurological, psychiatric and infectious diseases. However, glioprotective molecules can prevent or improve these glial dysfunctions, representing glial cells-targeting therapies. Therefore, this review will provide a brief summary of types and functions of glial cells and point out cellular and molecular mechanisms associated with gliotoxicity and glioprotection, potential glioprotective molecules and their mechanisms, as well as gliotherapy. In summary, we expect to address the relevance of gliotoxicity and glioprotection in the CNS homeostasis and diseases.
Collapse
Affiliation(s)
- André Quincozes-Santos
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.
- Programa de Pós-Graduação Em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.
| | - Camila Leite Santos
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Rômulo Rodrigo de Souza Almeida
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Amanda da Silva
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Natalie K Thomaz
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Naithan Ludian Fernandes Costa
- Programa de Pós-Graduação Em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Becker Weber
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Izaviany Schmitz
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Lara Scopel Medeiros
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Lívia Medeiros
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Bethina Segabinazzi Dotto
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Filipe Renato Pereira Dias
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Vanessa Sovrani
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
10
|
Human astrocytes and astrocytoma respond differently to resveratrol. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 37:102441. [PMID: 34302989 DOI: 10.1016/j.nano.2021.102441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/01/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022]
Abstract
A fundamental problem in oncology is that anticancer chemotherapeutics kill both cancer and healthy cells in the surrounding tissues. Resveratrol is a natural antioxidant with intriguing and opposing biological properties: it reduces viability of some cancer cells but not of non-transformed ones (in equimolar concentrations). Therefore, we examined resveratrol in human non-transformed primary astrocytes and astrocytoma. Resveratrol reduced reactive oxygen species in astrocytes, but not in astrocytoma. Such cell-type dependent response is particularly evident with analyses at the single cell level showing clear population difference in high and low glutathione levels. Due to resveratrol's poor aqueous solubility that limits its use in clinics, we incorporated it into stimulus-responsive micelles assembled from miktoarm polymers. This could be an attractive chemotherapeutic delivery strategy in nano-oncology. As a proof of principle, we show that these formulations containing resveratrol markedly decrease astrocytoma viability, particularly in combination with temozolomide, a first line chemotherapeutic for astrocytoma.
Collapse
|
11
|
Psenicka MW, Smith BC, Tinkey RA, Williams JL. Connecting Neuroinflammation and Neurodegeneration in Multiple Sclerosis: Are Oligodendrocyte Precursor Cells a Nexus of Disease? Front Cell Neurosci 2021; 15:654284. [PMID: 34234647 PMCID: PMC8255483 DOI: 10.3389/fncel.2021.654284] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
The pathology in neurodegenerative diseases is often accompanied by inflammation. It is well-known that many cells within the central nervous system (CNS) also contribute to ongoing neuroinflammation, which can promote neurodegeneration. Multiple sclerosis (MS) is both an inflammatory and neurodegenerative disease in which there is a complex interplay between resident CNS cells to mediate myelin and axonal damage, and this communication network can vary depending on the subtype and chronicity of disease. Oligodendrocytes, the myelinating cell of the CNS, and their precursors, oligodendrocyte precursor cells (OPCs), are often thought of as the targets of autoimmune pathology during MS and in several animal models of MS; however, there is emerging evidence that OPCs actively contribute to inflammation that directly and indirectly contributes to neurodegeneration. Here we discuss several contributors to MS disease progression starting with lesion pathology and murine models amenable to studying particular aspects of disease. We then review how OPCs themselves can play an active role in promoting neuroinflammation and neurodegeneration, and how other resident CNS cells including microglia, astrocytes, and neurons can impact OPC function. Further, we outline the very complex and pleiotropic role(s) of several inflammatory cytokines and other secreted factors classically described as solely deleterious during MS and its animal models, but in fact, have many neuroprotective functions and promote a return to homeostasis, in part via modulation of OPC function. Finally, since MS affects patients from the onset of disease throughout their lifespan, we discuss the impact of aging on OPC function and CNS recovery. It is becoming clear that OPCs are not simply a bystander during MS progression and uncovering the active roles they play during different stages of disease will help uncover potential new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Morgan W. Psenicka
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Brandon C. Smith
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - Rachel A. Tinkey
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Jessica L. Williams
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Brain Health Research Institute, Kent State University, Kent, OH, United States
| |
Collapse
|
12
|
Oxidized phosphatidylcholines found in multiple sclerosis lesions mediate neurodegeneration and are neutralized by microglia. Nat Neurosci 2021; 24:489-503. [PMID: 33603230 DOI: 10.1038/s41593-021-00801-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 01/12/2021] [Indexed: 01/30/2023]
Abstract
Neurodegeneration occurring in multiple sclerosis (MS) contributes to the progression of disability. It is therefore important to identify and neutralize the mechanisms that promote neurodegeneration in MS. Here, we report that oxidized phosphatidylcholines (OxPCs) found in MS lesions, previously identified as end-product markers of oxidative stress, are potent drivers of neurodegeneration. Cultured neurons and oligodendrocytes were killed by OxPCs, and this was ameliorated by microglia. After OxPC injection, mouse spinal cords developed focal demyelinating lesions with prominent axonal loss. The depletion of microglia that accumulated in OxPC lesions exacerbated neurodegeneration. Single-cell RNA sequencing of lesioned spinal cords identified unique subsets of TREM2high mouse microglia responding to OxPC deposition. TREM2 was detected in human MS lesions, and TREM2-/- mice exhibited worsened OxPC lesions. These results identify OxPCs as potent neurotoxins and suggest that enhancing microglia-mediated OxPC clearance via TREM2 could help prevent neurodegeneration in MS.
Collapse
|
13
|
N Kolodkin A, Sharma RP, Colangelo AM, Ignatenko A, Martorana F, Jennen D, Briedé JJ, Brady N, Barberis M, Mondeel TDGA, Papa M, Kumar V, Peters B, Skupin A, Alberghina L, Balling R, Westerhoff HV. ROS networks: designs, aging, Parkinson's disease and precision therapies. NPJ Syst Biol Appl 2020; 6:34. [PMID: 33106503 PMCID: PMC7589522 DOI: 10.1038/s41540-020-00150-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 08/28/2020] [Indexed: 12/11/2022] Open
Abstract
How the network around ROS protects against oxidative stress and Parkinson's disease (PD), and how processes at the minutes timescale cause disease and aging after decades, remains enigmatic. Challenging whether the ROS network is as complex as it seems, we built a fairly comprehensive version thereof which we disentangled into a hierarchy of only five simpler subnetworks each delivering one type of robustness. The comprehensive dynamic model described in vitro data sets from two independent laboratories. Notwithstanding its five-fold robustness, it exhibited a relatively sudden breakdown, after some 80 years of virtually steady performance: it predicted aging. PD-related conditions such as lack of DJ-1 protein or increased α-synuclein accelerated the collapse, while antioxidants or caffeine retarded it. Introducing a new concept (aging-time-control coefficient), we found that as many as 25 out of 57 molecular processes controlled aging. We identified new targets for "life-extending interventions": mitochondrial synthesis, KEAP1 degradation, and p62 metabolism.
Collapse
Affiliation(s)
- Alexey N Kolodkin
- Infrastructure for Systems Biology Europe (ISBE.NL), Amsterdam, The Netherlands.
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
- Molecular Cell Physiology, VU University Amsterdam, Amsterdam, The Netherlands.
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands.
| | - Raju Prasad Sharma
- Molecular Cell Physiology, VU University Amsterdam, Amsterdam, The Netherlands
- Environmental Engineering Laboratory, Departament d'Enginyeria Quimica, Universitat Rovira i Virgili, Tarragona, Spain
| | - Anna Maria Colangelo
- Infrastructure for Systems Biology Europe (ISBE.IT), Milan, Italy
- SysBio Centre of Systems Biology (ISBE.IT), University of Milano-Bicocca, Milan, Italy
- Laboratory of Neuroscience "R Levi-Montalcini" Dept of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Andrew Ignatenko
- Luxembourg Institute of Science and Technology (LIST), Esch-sur-Alzette, Luxembourg
| | - Francesca Martorana
- Infrastructure for Systems Biology Europe (ISBE.IT), Milan, Italy
- SysBio Centre of Systems Biology (ISBE.IT), University of Milano-Bicocca, Milan, Italy
- Laboratory of Neuroscience "R Levi-Montalcini" Dept of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Danyel Jennen
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Jacco J Briedé
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Nathan Brady
- Department of Molecular Microbiology & Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Surrey, UK
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Surrey, UK
| | - Thierry D G A Mondeel
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Surrey, UK
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Surrey, UK
| | - Michele Papa
- SysBio Centre of Systems Biology (ISBE.IT), University of Milano-Bicocca, Milan, Italy
- Infrastructure for Systems Biology Europe (ISBE.IT), Naples, Italy
- Department of Mental and Physical Health, University of Campania-L. Vanvitelli, Napoli, Italia
| | - Vikas Kumar
- Environmental Engineering Laboratory, Departament d'Enginyeria Quimica, Universitat Rovira i Virgili, Tarragona, Spain
- IISPV, Hospital Universitari Sant Joan de Reus, Universitat Rovira I Virgili, Reus, Spain
| | - Bernhard Peters
- Faculty of Science, Technology and Communication, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Lilia Alberghina
- Infrastructure for Systems Biology Europe (ISBE.IT), Milan, Italy
- SysBio Centre of Systems Biology (ISBE.IT), University of Milano-Bicocca, Milan, Italy
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Hans V Westerhoff
- Infrastructure for Systems Biology Europe (ISBE.NL), Amsterdam, The Netherlands.
- Molecular Cell Physiology, VU University Amsterdam, Amsterdam, The Netherlands.
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands.
- Manchester Centre for Integrative Systems Biology, School for Chemical Engineering and Analytical Science, The University of Manchester, Manchester, UK.
| |
Collapse
|
14
|
Structural and chemical changes in glial cells in the rat neocortex induced by constant occlusion of the middle cerebral artery. Acta Histochem 2020; 122:151573. [PMID: 32622419 DOI: 10.1016/j.acthis.2020.151573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/09/2020] [Accepted: 06/04/2020] [Indexed: 12/21/2022]
Abstract
Stroke-induced changes in neuroglia determine the basic conditions for the survival and damage of neurons in the ischemic core. Here, we studied the immunolocalization of glial cell line-derived neurotrophic factor (GDNF), glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor molecule 1 (Iba-1), and S-100β in the rat parietal cortex after constant occlusion of the middle cerebral artery. These cytoplasmic proteins are specific for different glial cell types. They are used as indicators of activated microglia and astrocytes in immunocytochemical studies. The distribution pattern of all markers changed dramatically with time. GFAP- and S-100β-positive astrocytes were observed in the penumbra zone and marked its boundaries. In days 1-8 after surgery, in the ischemic core, the number of S-100β-immunoreactive astrocytes decreased, and individual pyramidal cells appeared. S-100β-expressing pyramidal cells were localized in cortical layers III and V. They were only found in the ischemic core. Their proportion to the total number of cells was 37.3 ± 3.9 %, 22.2 ± 1.2 %, 16.3 ± 2.3 %, and 5.4 ± 0.3 % on days 1, 3, 8, and 14 after surgery. On day 21, no S-100β-expressing pyramidal cells were observed. The spatial density of GFAP- and S-100β-positive astrocytes increased in the penumbra region adjacent to the ischemic core and decreased in the penumbral periphery. As a result, the width of the perifocal penumbra zone decreased substantially at later stages of the stroke. In the penumbra, on days 1-3 after ischemic injury, GDNF immunoreactivity was mainly localized in neurons, while later on (days 8-21) it was mainly constrained to astrocyte glia. In intact rats, GDNF-positive neurons were situated in cortical layers II/III and V/VI and made up 52 ± 4.5 % of the total neuron population. Their proportion to the total number of neurons was 29 ± 2.1 %, 13.8 ± 0.6 %, and 3.1 ± 0.2 % on days 1, 8, and 21 after surgery. The number of GDNF-positive astrocytes, on the opposite, increased with time after ischemic injury. Iba-1-reactive microglia was mainly localized to the ischemic core. Microglial cells appeared activated as evidenced by their increased size and decreased number of processes and branching density. The spatial density of microglia reached a peak on day 8 after ischemic injury both in the ischemic core and penumbra. An increase in the number of Iba-1-reactive microglia in the ischemic core correlated with a decrease of the number of GFAP-positive astrocytes. The results are discussed in the context of participation of neuroglia in regulation of various neuroprotective and destructive processes.
Collapse
|
15
|
Duong P, Tenkorang MAA, Trieu J, McCuiston C, Rybalchenko N, Cunningham RL. Neuroprotective and neurotoxic outcomes of androgens and estrogens in an oxidative stress environment. Biol Sex Differ 2020; 11:12. [PMID: 32223745 PMCID: PMC7104511 DOI: 10.1186/s13293-020-0283-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/20/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The role of sex hormones on cellular function is unclear. Studies show androgens and estrogens are protective in the CNS, whereas other studies found no effects or damaging effects. Furthermore, sex differences have been observed in multiple oxidative stress-associated CNS disorders, such as Alzheimer's disease, depression, and Parkinson's disease. The goal of this study is to examine the relationship between sex hormones (i.e., androgens and estrogens) and oxidative stress on cell viability. METHODS N27 and PC12 neuronal and C6 glial phenotypic cell lines were used. N27 cells are female rat derived, whereas PC12 cells and C6 cells are male rat derived. These cells express estrogen receptors and the membrane-associated androgen receptor variant, AR45, but not the full-length androgen receptor. N27, PC12, and C6 cells were exposed to sex hormones either before or after an oxidative stressor to examine neuroprotective and neurotoxic properties, respectively. Estrogen receptor and androgen receptor inhibitors were used to determine the mechanisms mediating hormone-oxidative stress interactions on cell viability. Since the presence of AR45 in the human brain tissue was unknown, we examined the postmortem brain tissue from men and women for AR45 protein expression. RESULTS Neither androgens nor estrogens were protective against subsequent oxidative stress insults in glial cells. However, these hormones exhibited neuroprotective properties in neuronal N27 and PC12 cells via the estrogen receptor. Interestingly, a window of opportunity exists for sex hormone neuroprotection, wherein temporary hormone deprivation blocked neuroprotection by sex hormones. However, if sex hormones are applied following an oxidative stressor, they exacerbated oxidative stress-induced cell loss in neuronal and glial cells. CONCLUSIONS Sex hormone action on cell viability is dependent on the cellular environment. In healthy neuronal cells, sex hormones are protective against oxidative stress insults via the estrogen receptor, regardless of sex chromosome complement (XX, XY). However, in unhealthy (e.g., high oxidative stress) cells, sex hormones exacerbated oxidative stress-induced cell loss, regardless of cell type or sex chromosome complement. The non-genomic AR45 receptor, which is present in humans, mediated androgen's damaging effects, but it is unknown which receptor mediated estrogen's damaging effects. These differential effects of sex hormones that are dependent on the cellular environment, receptor profile, and cell type may mediate the observed sex differences in oxidative stress-associated CNS disorders.
Collapse
Affiliation(s)
- Phong Duong
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Mavis A A Tenkorang
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Jenny Trieu
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Clayton McCuiston
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Nataliya Rybalchenko
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Rebecca L Cunningham
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA. .,Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, 3400 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA.
| |
Collapse
|
16
|
Peynshaert K, Devoldere J, Philips F, Vergauwe F, De Smedt S, Remaut K. Influence of pathogenic stimuli on Müller cell transfection by lipoplexes. Eur J Pharm Biopharm 2020; 150:87-95. [PMID: 32173604 DOI: 10.1016/j.ejpb.2020.03.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 02/14/2020] [Accepted: 03/09/2020] [Indexed: 11/24/2022]
Abstract
Neuroprotection is a mutation-independent therapeutic strategy that seeks to enhance the survival of neuronal cell types through delivery of neuroprotective factors. The Müller cell, a retinal glial cell type appreciated for its unique morphology and neuroprotective functions, could be regarded as an ideal target for this strategy by functioning as a secretion platform within the retina following uptake of a transgene of our choice. In this in vitro study we aimed to investigate the capability of Müller cells to take up a standard liposomal vector (i.e. Lipofectamine 2000) and process its pDNA or mRNA cargo into the reporter GFP protein. By doing so, we found that mRNA outperformed pDNA in Müller cell transfection efficiency. Since neuroprotection is explored as a therapy for diabetic retinopathy and glaucoma, we furthermore examined the Müller cell's lipoplex-induced transfection efficiency and cytotoxicity in stressful conditions linked to these diseases - i.e. hypoxia, hyperglycemia and oxidative stress. Interestingly, Müller cells were able of maintaining high GFP expression regardless of these noxious stimuli. In terms of lipoplex-induced toxicity, hyperglycemia seemed to have a protective effect while hypoxia and oxidative stress led to a slightly higher toxicity. In conclusion, our study indicates that mRNA-lipoplexes have potential in transfecting Müller cells in healthy as well as diseased conditions.
Collapse
Affiliation(s)
- Karen Peynshaert
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Joke Devoldere
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Frederik Philips
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Fauve Vergauwe
- Odisee University College, Technology Campus Ghent, Gebroeders De Smetstraat 1, 9000 Ghent, Belgium
| | - Stefaan De Smedt
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Katrien Remaut
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
17
|
Zambuto SG, Serrano JF, Vilbert AC, Lu Y, Harley BAC, Pedron S. Response of neuroglia to hypoxia-induced oxidative stress using enzymatically crosslinked hydrogels. MRS COMMUNICATIONS 2020; 10:83-90. [PMID: 32719734 PMCID: PMC7384750 DOI: 10.1557/mrc.2019.159] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/04/2019] [Indexed: 05/09/2023]
Abstract
Three-dimensional cultures have exciting potential to mimic aspects of healthy and diseased brain tissue to examine the role of physiological conditions on neural biomarkers, as well as disease onset and progression. Hypoxia is associated with oxidative stress, mitochondrial damage, and inflammation, key processes potentially involved in Alzheimer's and multiple sclerosis. We describe the use of an enzymatically-crosslinkable gelatin hydrogel system within a microfluidic device to explore the effects of hypoxia-induced oxidative stress on rat neuroglia, human astrocyte reactivity, and myelin production. This versatile platform offers new possibilities for drug discovery and modeling disease progression.
Collapse
Affiliation(s)
- Samantha G Zambuto
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 1406 W. Green St, Urbana, IL 61801, USA
| | - Julio F Serrano
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, IL 61801, USA; Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, IL 61801, USA
| | - Avery C Vilbert
- Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99354, USA
| | - Yi Lu
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, IL 61801, USA; Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99354, USA; Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL 61801, USA
| | - Brendan A C Harley
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, IL 61801, USA; Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, IL 61801, USA
| | - Sara Pedron
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, IL 61801, USA; Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, IL 61801, USA
| |
Collapse
|
18
|
Soares MSP, Pedra NS, Bona NP, de Souza AÁ, Teixeira FC, Azambuja JH, Wyse AT, Braganhol E, Stefanello FM, Spanevello RM. Methionine and methionine sulfoxide induces neurochemical and morphological changes in cultured astrocytes: Involvement of Na +, K +-ATPase activity, oxidative status, and cholinergic and purinergic signaling. Neurotoxicology 2019; 77:60-70. [PMID: 31883448 DOI: 10.1016/j.neuro.2019.12.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/18/2019] [Accepted: 12/21/2019] [Indexed: 12/25/2022]
Abstract
Hypermethioninemia is an inherited metabolic disorder characterized by high concentration of methionine (Met) and its metabolites such as methionine sulfoxide (Met-SO), which may lead to development of neurological alterations. The aim of this study was to investigate the in vitro effects of Met or Met-SO on viability, proliferation, morphology, and neurochemical parameters in primary culture of cortical astrocytes, after treatment with 1 or 2 mM Met or 0.5 mM Met-SO, for 24, 48, and 72 h. Met or Met-SO did not affect cell viability and proliferation but induced astrocyte hypertrophy. Acetylcholinesterase activity was increased, while Na+, K+-ATPase activity was decreased by 2 mM Met, Met-SO, or Met (1 and 2 mM) + Met-SO (P < 0.05). ATP and AMP hydrolysis was decreased by Met (1 and 2 mM), Met-SO and Met (1 and 2 mM) + Met-SO treatment, while ADP hydrolysis was enhanced by Met-SO and Met (1 and 2 mM) + Met-SO (P < 0.05). Superoxide dismutase activity was increased by Met-SO and Met (1 and 2 mM) + Met-SO (P < 0.05). Catalase and glutathione S-transferase activities were reduced by Met or Met-SO treatment for 48 and 72 h (P < 0.05). Reactive oxygen species and total thiol content was reduced by Met or Met-SO treatment for 24, 48, and 72 h while nitrite and thiobarbituric acid reactive substance levels were increased under the same experimental conditions (P < 0.05). High concentrations of Met and Met-SO do not cause cell death but induced changes in astrocyte function. These alterations in astrocytic homeostasis may be associated with neurological symptoms found in hypermethioninemia.
Collapse
Affiliation(s)
- Mayara Sandrielly Pereira Soares
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil.
| | - Nathalia Stark Pedra
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Natália Pontes Bona
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Anita Ávila de Souza
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Fernanda Cardoso Teixeira
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Juliana Hofstatter Azambuja
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Angela Ts Wyse
- Laboratório de Neuroproteção e Doença Metabólica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Elizandra Braganhol
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Francieli Moro Stefanello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Roselia Maria Spanevello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| |
Collapse
|
19
|
Ali AM, Kunugi H. Bee honey protects astrocytes against oxidative stress: A preliminary in vitro investigation. Neuropsychopharmacol Rep 2019; 39:312-314. [PMID: 31529692 PMCID: PMC7292328 DOI: 10.1002/npr2.12079] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/31/2019] [Accepted: 08/22/2019] [Indexed: 01/10/2023] Open
Abstract
Aim Bee honey is widely used as a bioactive food to improve general health and produce therapeutic benefits in various physical disorders. It also improves cognitive and mood‐related behaviors and symptoms in mice and humans. Still, its direct effect on brain cells is unclear. Here, we examined the effect of whole honey on the survival of astrocytes exposed to oxidative stress. Methods Cultured cortical astrocytes were treated with honey (0.1%, 0.3%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 3%, and 5% [v/v]) for 24 hours followed by H2O2 (100 μmol/L) for 3 hours. Cellular viability was assessed with MTT assay. Results Honey prevented cellular death in a dose‐dependent manner compared with H2O2‐treated cells. Honey at 1% concentration had the most significant effect (P = .015). Conclusion Bee honey exerts a neuroprotective effect through its antioxidant activity. This study tested the effect of bee honey on the survival of astrocytes exposed to H2O2‐induced oxidative stress. Honey prevented cellular death in a dose‐dependent manner compared with H2O2‐treated cells. Honey at 1% concentration had the most significant effect (P = .015).![]()
Collapse
Affiliation(s)
- Amira Mohammed Ali
- Department of Mental Disorder Research, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Tokyo, Japan.,Department of Psychiatric Nursing and Mental Health, Faculty of Nursing, Alexandria University, Alexandria, Egypt
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Tokyo, Japan
| |
Collapse
|