1
|
Cai J, Chen Y, She Y, He X, Feng H, Sun H, Yin M, Gao J, Sheng C, Li Q, Xiao M. Aerobic exercise improves astrocyte mitochondrial quality and transfer to neurons in a mouse model of Alzheimer's disease. Brain Pathol 2025; 35:e13316. [PMID: 39462160 PMCID: PMC11961210 DOI: 10.1111/bpa.13316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024] Open
Abstract
Mitochondrial dysfunction is a well-established hallmark of Alzheimer's disease (AD). Despite recent documentation of transcellular mitochondrial transfer, its role in the pathogenesis of AD remains unclear. In this study, we report an impairment of mitochondrial quality within the astrocytes and neurons of adult 5 × FAD mice. Following treatment with mitochondria isolated from aged astrocytes induced by exposure to amyloid protein or extended cultivation, cultured neurons exhibited an excessive generation of reactive oxygen species and underwent neurite atrophy. Notably, aerobic exercise enhanced mitochondrial quality by upregulating CD38 within hippocampal astrocytes of 5 × FAD mice. Conversely, the knockdown of CD38 diminished astrocytic-neuronal mitochondrial transfer, thereby abolishing the ameliorative effects of aerobic exercise on neuronal oxidative stress, β-amyloid plaque deposition, and cognitive dysfunction in 5 × FAD mice. These findings unveil an unexpected mechanism through which aerobic exercise facilitates the transference of healthy mitochondria from astrocytes to neurons, thus countering the AD-like progression.
Collapse
Affiliation(s)
- Jiachen Cai
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Nanjing Brain Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Yan Chen
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Nanjing Brain Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Yuzhu She
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Nanjing Brain Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Xiaoxin He
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Nanjing Brain Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Hu Feng
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
| | - Huaiqing Sun
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Department of NeurologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Mengmei Yin
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Department of NeurologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Junying Gao
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Department of AnatomyNanjing Medical UniversityNanjingChina
| | - Chengyu Sheng
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
| | - Qian Li
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Nanjing Brain Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Ming Xiao
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Nanjing Brain Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| |
Collapse
|
2
|
Schröder S, Sakib MS, Krüger DM, Pena T, Burkhardt S, Schütz AL, Sananbenesi F, Fischer A. LncRNA 3222401L13Rik Is Upregulated in Aging Astrocytes and Regulates Neuronal Support Function Through Interaction with Npas3. Noncoding RNA 2025; 11:2. [PMID: 39846680 PMCID: PMC11755665 DOI: 10.3390/ncrna11010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
Aging leads to cognitive decline and increased risk of neurodegenerative diseases. While molecular changes in central nervous system (CNS) cells contribute to this decline, the mechanisms are not fully understood. Long non-coding RNAs (lncRNAs) are key regulators of cellular functions. Background/Objectives: The roles of lncRNAs in aging, especially in glial cells, are not well characterized. Methods: We investigated lncRNA expression in non-neuronal cells from aged mice and identified 3222401L13Rik, a previously unstudied lncRNA, as upregulated in astrocytes during aging. Results: Knockdown of 3222401L13Rik in primary astrocytes revealed its critical role in regulating genes for neuronal support and synapse organization, a function conserved in human iPSC-derived astrocytes. A 3222401L13Rik interacts with the transcription factor Neuronal PAS Domain Protein 3 (Npas3), and overexpression of Npas3 rescues deficits in astrocytes lacking 3222401L13Rik. Conclusions: These data suggest that 3222401L13Rik upregulation may help delay age-related cognitive decline.
Collapse
Affiliation(s)
- Sophie Schröder
- Department for Systems Medicine and Epigenetics, German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany; (S.S.); (M.S.S.); (D.M.K.); (T.P.); (S.B.)
| | - M. Sadman Sakib
- Department for Systems Medicine and Epigenetics, German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany; (S.S.); (M.S.S.); (D.M.K.); (T.P.); (S.B.)
| | - Dennis M. Krüger
- Department for Systems Medicine and Epigenetics, German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany; (S.S.); (M.S.S.); (D.M.K.); (T.P.); (S.B.)
- Bioinformatics Unit, German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany
- Research Group for Genome Dynamics in Brain Diseases, German Center for Neurodegenerative Diseases, 37075 Göttingen, Germany; (A.-L.S.); (F.S.)
| | - Tonatiuh Pena
- Department for Systems Medicine and Epigenetics, German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany; (S.S.); (M.S.S.); (D.M.K.); (T.P.); (S.B.)
- Bioinformatics Unit, German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany
| | - Susanne Burkhardt
- Department for Systems Medicine and Epigenetics, German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany; (S.S.); (M.S.S.); (D.M.K.); (T.P.); (S.B.)
- Research Group for Genome Dynamics in Brain Diseases, German Center for Neurodegenerative Diseases, 37075 Göttingen, Germany; (A.-L.S.); (F.S.)
| | - Anna-Lena Schütz
- Research Group for Genome Dynamics in Brain Diseases, German Center for Neurodegenerative Diseases, 37075 Göttingen, Germany; (A.-L.S.); (F.S.)
| | - Farahnaz Sananbenesi
- Research Group for Genome Dynamics in Brain Diseases, German Center for Neurodegenerative Diseases, 37075 Göttingen, Germany; (A.-L.S.); (F.S.)
| | - André Fischer
- Department for Systems Medicine and Epigenetics, German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany; (S.S.); (M.S.S.); (D.M.K.); (T.P.); (S.B.)
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, 37075 Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
3
|
Pérez LA, Palacios E, González MF, Leyton-Rivera I, Martínez-Meza S, Pérez-Núñez R, Jeldes E, Avalos AM, Díaz J, Leyton L. A Pro-Inflammatory Stimulus versus Extensive Passaging of DITNC1 Astrocyte Cultures as Models to Study Astrogliosis. Int J Mol Sci 2024; 25:9454. [PMID: 39273404 PMCID: PMC11394751 DOI: 10.3390/ijms25179454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Astrogliosis is a process by which astrocytes, when exposed to inflammation, exhibit hypertrophy, motility, and elevated expression of reactivity markers such as Glial Fibrillar Acidic Protein, Vimentin, and Connexin43. Since 1999, our laboratory in Chile has been studying molecular signaling pathways associated with "gliosis" and has reported that reactive astrocytes upregulate Syndecan 4 and αVβ3 Integrin, which are receptors for the neuronal glycoprotein Thy-1. Thy-1 engagement stimulates adhesion and migration of reactive astrocytes and induces neurons to retract neurites, thus hindering neuronal network repair. Reportedly, we have used DITNC1 astrocytes and neuron-like CAD cells to study signaling mechanisms activated by the Syndecan 4-αVβ3 Integrin/Thy-1 interaction. Importantly, the sole overexpression of β3 Integrin in non-reactive astrocytes turns them into reactive cells. In vitro, extensive passaging is a simile for "aging", and aged fibroblasts have shown β3 Integrin upregulation. However, it is not known if astrocytes upregulate β3 Integrin after successive cell passages. Here, we hypothesized that astrocytes undergoing long-term passaging increase β3 Integrin expression levels and behave as reactive astrocytes without needing pro-inflammatory stimuli. We used DITNC1 cells with different passage numbers to study reactivity markers using immunoblots, immunofluorescence, and astrocyte adhesion/migration assays. We also evaluated β3 Integrin levels by immunoblot and flow cytometry, as well as the neurotoxic effects of reactive astrocytes. Serial cell passaging mimicked the effects of inflammatory stimuli, inducing astrocyte reactivity. Indeed, in response to Thy-1, β3 Integrin levels, as well as cell adhesion and migration, gradually increased with multiple passages. Importantly, these long-lived astrocytes expressed and secreted factors that inhibited neurite outgrowth and caused neuronal death, just like reactive astrocytes in culture. Therefore, we describe two DITNC1 cell types: a non-reactive type that can be activated with Tumor Necrosis Factor (TNF) and another one that exhibits reactive astrocyte features even in the absence of TNF treatment. Our results emphasize the importance of passage numbers in cell behavior. Likewise, we compare the pro-inflammatory stimulus versus long-term in-plate passaging of cell cultures and introduce them as astrocyte models to study the reactivity process.
Collapse
Affiliation(s)
- Leonardo A Pérez
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| | - Esteban Palacios
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Laboratorio de Microbiología Celular, Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 833-0546, Chile
| | - María Fernanda González
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| | - Ignacio Leyton-Rivera
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| | - Samuel Martínez-Meza
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| | - Ramón Pérez-Núñez
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| | - Emanuel Jeldes
- Andes Biotechnologies SpA, Santiago 7750000, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia y Vida, Santiago 7750000, Chile
| | - Ana María Avalos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 7500912, Chile
| | - Jorge Díaz
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| |
Collapse
|
4
|
Giovannuzzi S, Chavarria D, Provensi G, Leri M, Bucciantini M, Carradori S, Bonardi A, Gratteri P, Borges F, Nocentini A, Supuran CT. Dual Inhibitors of Brain Carbonic Anhydrases and Monoamine Oxidase-B Efficiently Protect against Amyloid-β-Induced Neuronal Toxicity, Oxidative Stress, and Mitochondrial Dysfunction. J Med Chem 2024; 67:4170-4193. [PMID: 38436571 DOI: 10.1021/acs.jmedchem.4c00045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
We report here the first dual inhibitors of brain carbonic anhydrases (CAs) and monoamine oxidase-B (MAO-B) for the management of Alzheimer's disease. Classical CA inhibitors (CAIs) such as methazolamide prevent amyloid-β-peptide (Aβ)-induced overproduction of reactive oxygen species (ROS) and mitochondrial dysfunction. MAO-B is also implicated in ROS production, cholinergic system disruption, and amyloid plaque formation. In this work, we combined a reversible MAO-B inhibitor of the coumarin and chromone type with benzenesulfonamide fragments as highly effective CAIs. A hit-to-lead optimization led to a significant set of derivatives showing potent low nanomolar inhibition of the target brain CAs (KIs in the range of 0.1-90.0 nM) and MAO-B (IC50 in the range of 6.7-32.6 nM). Computational studies were conducted to elucidate the structure-activity relationship and predict ADMET properties. The most effective multitarget compounds totally prevented Aβ-related toxicity, reverted ROS formation, and restored the mitochondrial functionality in an SH-SY5Y cell model surpassing the efficacy of single-target drugs.
Collapse
Affiliation(s)
- Simone Giovannuzzi
- NEUROFARBA Department, Pharmaceutical and Nutraceutical Section, University of Florence, Via U. Schiff 6, Sesto Fiorentino, 50019 Florence, Italy
| | - Daniel Chavarria
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Gustavo Provensi
- NEUROFARBA Department, Section of Pharmacology and Toxicology, University of Florence, via G. Pieraccini 6, 50139 Florence, Italy
| | - Manuela Leri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Monica Bucciantini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Simone Carradori
- Department of Pharmacy, "G. D'Annunzio" University of Chieti and Pescara, via dei Vestini 31, 66100 Chieti, Italy
| | - Alessandro Bonardi
- NEUROFARBA Department, Pharmaceutical and Nutraceutical Section, Laboratory of Molecular Modeling Cheminformatics & QSAR, University of Florence, Via U. Schiff 6, Sesto Fiorentino, 50019 Florence, Italy
| | - Paola Gratteri
- NEUROFARBA Department, Pharmaceutical and Nutraceutical Section, Laboratory of Molecular Modeling Cheminformatics & QSAR, University of Florence, Via U. Schiff 6, Sesto Fiorentino, 50019 Florence, Italy
| | - Fernanda Borges
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Alessio Nocentini
- NEUROFARBA Department, Pharmaceutical and Nutraceutical Section, University of Florence, Via U. Schiff 6, Sesto Fiorentino, 50019 Florence, Italy
| | - Claudiu T Supuran
- NEUROFARBA Department, Pharmaceutical and Nutraceutical Section, University of Florence, Via U. Schiff 6, Sesto Fiorentino, 50019 Florence, Italy
| |
Collapse
|
5
|
Lin X, Guo Z, Lin S, Qiu Y. Transcriptional expression of radiation-induced early cortical morphological alterations and its association with radiation necrosis in patients with nasopharyngeal carcinoma. Radiother Oncol 2023; 186:109770. [PMID: 37385380 DOI: 10.1016/j.radonc.2023.109770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023]
Abstract
PURPOSE To explore the effects of standard radiotherapy on cortical morphology and its potential transcriptional expression, and to determine the predictive power of cortical morphological measurement at the early stage for radiation necrosis (RN) occurrence within 3 years post-radiotherapy in patients with nasopharyngeal carcinoma (NPC). METHODS 185 NPC patients participated. Pre-treatment and post-radiotherapy (1-3 months) structural MRI were collected longitudinally and prospectively. Multiple cortical morphological indices were compared between pre-treatment and post-radiotherapy. Brain-wide gene expression was used to assess the transcriptional profiles associated with radiation-induced cortical morphological changes. Machine learning was used to construct predictive models for RN with cortical morphological alterations at the early stage. RESULTS Relative to pre-treatment, NPC patients exhibited a widespread reduction in cortical volume (CV) and cortical thickness (CT) post-radiotherapy (p < 0.001). Partial least squares regression analysis revealed that radiotherapy-related cortical atrophy was closely related to transcriptional profiles (p < 0.001), with the most correlated genes enriched in ATPase Na+/K+ transporting alpha-1 and alpha-3 polypeptide and respiratory electron transport chain. Furthermore, models constructed with cortical morphological features at 1-3 months post-radiotherapy had favorable predictive power for RN occurrence in NPC patients within 3-year follow-up, the area under the curve was 0.854 and 0.843 for CV and CT, respectively. CONCLUSIONS NPC patients exhibited widespread cortical atrophy at 1-3 months post-radiotherapy, which was closely correlated with dysfunction of the ATPase Na+/K+ transporting alpha-1 and alpha-3 polypeptide and respiratory electron transport chain. Cortical morphology at 1-3 months post-radiotherapy may serve as an early biomarker for identifying RN.
Collapse
Affiliation(s)
- Xiaoshan Lin
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, 89 Taoyuan road, Nanshan district, Shenzhen 518052, China
| | - Zheng Guo
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen 518055, China
| | - Shiwei Lin
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, 89 Taoyuan road, Nanshan district, Shenzhen 518052, China
| | - Yingwei Qiu
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, 89 Taoyuan road, Nanshan district, Shenzhen 518052, China.
| |
Collapse
|
6
|
Fan J, Miao Y, Zhao Y, Guan Y, Zhang L, Pan L, Feng Q, Yao J, Sun C. Icaritin inhibits oxidative stress in murine astrocytes by binding to Orai1 to block store-operated calcium channel. Chem Biol Drug Des 2023; 101:873-882. [PMID: 36527176 DOI: 10.1111/cbdd.14193] [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: 10/13/2022] [Revised: 11/18/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Previous study has shown that icaritin (ICT) has meaningful protective effect on cerebral ischemic stroke, and this study aimed to investigate its mechanism from the aspect of protecting astrocytes from oxidative stress. Murine primary astrocytes were pretreated by ICT and exposed to H2 O2 to induce oxidative stress. The results indicated that ICT inhibited H2 O2 -induced astrocytes apoptosis, decreased Bax and cleaved caspase-3, and increased Bcl-2. In addition, ICT inhibited H2 O2 -induced oxidative stress, increased mitochondrial membrane potential (ΔΨm ), and maintained mitochondrial morphology. ICT decreased the synthesis of malondialdehyde and increased the activity of glutathione peroxidase, catalase, and superoxide dismutase. Moreover, ICT suppressed the transient and resting intracellular Ca2+ overload. Further investigation revealed that ICT could target the combination with Orai1 to block store-operated calcium channel induced by H2 O2 . However, ICT did not enhance the protective effect of RO2959, a selective blocker of Orai1. These results indicate that ICT can play a neuroprotective role against oxidative stress injury by binding to Orai1 to block SOCC.
Collapse
Affiliation(s)
- Jianwei Fan
- Lunan Pharmaceutical Group Co. Ltd., Linyi, China
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Yu Miao
- Lunan Pharmaceutical Group Co. Ltd., Linyi, China
| | - Yun Zhao
- Lunan Pharmaceutical Group Co. Ltd., Linyi, China
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Yongxia Guan
- Lunan Pharmaceutical Group Co. Ltd., Linyi, China
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Li Zhang
- Lunan Pharmaceutical Group Co. Ltd., Linyi, China
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Lihong Pan
- Lunan Pharmaceutical Group Co. Ltd., Linyi, China
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Qun Feng
- Lunan Pharmaceutical Group Co. Ltd., Linyi, China
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Jingchun Yao
- Lunan Pharmaceutical Group Co. Ltd., Linyi, China
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Chenghong Sun
- Lunan Pharmaceutical Group Co. Ltd., Linyi, China
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| |
Collapse
|
7
|
Villablanca C, Vidal R, Gonzalez-Billault C. Are cytoskeleton changes observed in astrocytes functionally linked to aging? Brain Res Bull 2023; 196:59-67. [PMID: 36935053 DOI: 10.1016/j.brainresbull.2023.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/22/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
Astrocytes are active participants in the performance of the Central Nervous System (CNS) in both health and disease. During aging, astrocytes are susceptible to reactive astrogliosis, a molecular state characterized by functional changes in response to pathological situations, and cellular senescence, characterized by loss of cell division, apoptosis resistance, and gain of proinflammatory functions. This results in two different states of astrocytes, which can produce proinflammatory phenotypes with harmful consequences in chronic conditions. Reactive astrocytes and senescent astrocytes share morpho-functional features that are dependent on the organization of the cytoskeleton. However, such changes in the cytoskeleton have yet to receive the necessary attention to explain their role in the alterations of astrocytes that are associated with aging and pathologies. In this review, we summarize all the available findings that connect changes in the cytoskeleton of the astrocytes with aging. In addition, we discuss future avenues that we believe will guide such a novel topic.
Collapse
Affiliation(s)
- Cristopher Villablanca
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - René Vidal
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Christian Gonzalez-Billault
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile; Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile; Institute for Nutrition and Food Technologies, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
8
|
Anderson G. Amyotrophic Lateral Sclerosis Pathoetiology and Pathophysiology: Roles of Astrocytes, Gut Microbiome, and Muscle Interactions via the Mitochondrial Melatonergic Pathway, with Disruption by Glyphosate-Based Herbicides. Int J Mol Sci 2022; 24:ijms24010587. [PMID: 36614029 PMCID: PMC9820185 DOI: 10.3390/ijms24010587] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
The pathoetiology and pathophysiology of motor neuron loss in amyotrophic lateral sclerosis (ALS) are still to be determined, with only a small percentage of ALS patients having a known genetic risk factor. The article looks to integrate wider bodies of data on the biological underpinnings of ALS, highlighting the integrative role of alterations in the mitochondrial melatonergic pathways and systemic factors regulating this pathway across a number of crucial hubs in ALS pathophysiology, namely glia, gut, and the muscle/neuromuscular junction. It is proposed that suppression of the mitochondrial melatonergic pathway underpins changes in muscle brain-derived neurotrophic factor, and its melatonergic pathway mimic, N-acetylserotonin, leading to a lack of metabolic trophic support at the neuromuscular junction. The attenuation of the melatonergic pathway in astrocytes prevents activation of toll-like receptor agonists-induced pro-inflammatory transcription factors, NF-kB, and yin yang 1, from having a built-in limitation on inflammatory induction that arises from their synchronized induction of melatonin release. Such maintained astrocyte activation, coupled with heightened microglia reactivity, is an important driver of motor neuron susceptibility in ALS. Two important systemic factors, gut dysbiosis/permeability and pineal melatonin mediate many of their beneficial effects via their capacity to upregulate the mitochondrial melatonergic pathway in central and systemic cells. The mitochondrial melatonergic pathway may be seen as a core aspect of cellular function, with its suppression increasing reactive oxygen species (ROS), leading to ROS-induced microRNAs, thereby altering the patterning of genes induced. It is proposed that the increased occupational risk of ALS in farmers, gardeners, and sportsmen and women is intimately linked to exposure, whilst being physically active, to the widely used glyphosate-based herbicides. This has numerous research and treatment implications.
Collapse
Affiliation(s)
- George Anderson
- CRC Scotland & London, Eccleston Square, London SW1V 1PG, UK
| |
Collapse
|
9
|
Toledano-Díaz A, Álvarez MI, Toledano A. The relationships between neuroglial and neuronal changes in Alzheimer's disease, and the related controversies II: gliotherapies and multimodal therapy. J Cent Nerv Syst Dis 2022; 14:11795735221123896. [PMID: 36407561 PMCID: PMC9666878 DOI: 10.1177/11795735221123896] [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: 08/18/2021] [Accepted: 08/05/2022] [Indexed: 08/30/2023] Open
Abstract
Since the original description of Alzheimer´s disease (AD), research into this condition has mainly focused on assessing the alterations to neurons associated with dementia, and those to the circuits in which they are involved. In most of the studies on human brains and in many models of AD, the glial cells accompanying these neurons undergo concomitant alterations that aggravate the course of neurodegeneration. As a result, these changes to neuroglial cells are now included in all the "pathogenic cascades" described in AD. Accordingly, astrogliosis and microgliosis, the main components of neuroinflammation, have been integrated into all the pathogenic theories of this disease, as discussed in this part of the two-part monograph that follows an accompanying article on gliopathogenesis and glioprotection. This initial reflection verified the implication of alterations to the neuroglia in AD, suggesting that these cells may also represent therapeutic targets to prevent neurodegeneration. In this second part of the monograph, we will analyze the possibilities of acting on glial cells to prevent or treat the neurodegeneration that is the hallmark of AD and other pathologies. Evidence of the potential of different pharmacological, non-pharmacological, cell and gene therapies (widely treated) to prevent or treat this disease is now forthcoming, in most cases as adjuncts to other therapies. A comprehensive AD multimodal therapy is proposed in which neuronal and neuroglial pharmacological treatments are jointly considered, as well as the use of new cell and gene therapies and non-pharmacological therapies that tend to slow down the progress of dementia.
Collapse
|
10
|
Feliziani C, Fernandez M, Quasollo G, Holstein D, Bairo SM, Paton JC, Paton AW, de Batista J, Lechleiter JD, Bollo M. Ca 2+ signalling system initiated by endoplasmic reticulum stress stimulates PERK activation. Cell Calcium 2022; 106:102622. [PMID: 35908318 PMCID: PMC9982837 DOI: 10.1016/j.ceca.2022.102622] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/11/2022] [Accepted: 07/05/2022] [Indexed: 01/25/2023]
Abstract
The accumulation of unfolded proteins within the Endoplasmic Reticulum (ER) activates a signal transduction pathway termed the unfolded protein response (UPR), which attempts to restore ER homoeostasis. If this cannot be done, UPR signalling ultimately induces apoptosis. Ca2+ depletion in the ER is a potent inducer of ER stress. Despite the ubiquity of Ca2+ as an intracellular messenger, the precise mechanism(s) by which Ca2+ release affects the UPR remains unknown. Tethering a genetically encoded Ca2+ indicator (GCamP6) to the ER membrane revealed novel Ca2+ signalling events initiated by Ca2+ microdomains in human astrocytes under ER stress, induced by tunicamycin (Tm), an N-glycosylation inhibitor, as well as in a cell model deficient in all three inositol triphosphate receptor isoforms. Pharmacological and molecular studies indicate that these local events are mediated by translocons and that the Ca2+ microdomains impact (PKR)-like-ER kinase (PERK), an UPR sensor, activation. These findings reveal the existence of a Ca2+ signal mechanism by which stressor-mediated Ca2+ release regulates ER stress.
Collapse
Affiliation(s)
- Constanza Feliziani
- Instituto de Investigación Médica M y M
Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, 2434 Friuli,
Córdoba 5016, Argentina
| | - Macarena Fernandez
- Instituto de Investigación Médica M y M
Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, 2434 Friuli,
Córdoba 5016, Argentina
| | - Gonzalo Quasollo
- Instituto de Investigación Médica M y M
Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, 2434 Friuli,
Córdoba 5016, Argentina
| | - Deborah Holstein
- Department of Cell Systems and Anatomy, UT Health San
Antonio, 8403 Floyd Curl Dr., San Antonio, TX 78229-3904, USA
| | - Sebastián M Bairo
- Instituto de Investigación Médica M y M
Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, 2434 Friuli,
Córdoba 5016, Argentina
| | - James C Paton
- Research Centre for Infectious Diseases, School of
Molecular and Biomedical Science, University of Adelaide, South Australia 5005,
Australia
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, School of
Molecular and Biomedical Science, University of Adelaide, South Australia 5005,
Australia
| | - Juan de Batista
- Instituto Universitario de Ciencias Biomédicas de
Córdoba (IUCBC), Hospital Privado Universitario de Córdoba, 420
Naciones Unidas, Córdoba 5016, Argentina
| | - James D Lechleiter
- Department of Cell Systems and Anatomy, UT Health San
Antonio, 8403 Floyd Curl Dr., San Antonio, TX 78229-3904, USA
| | - Mariana Bollo
- Instituto de Investigación Médica M y M Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, 2434 Friuli, Córdoba 5016, Argentina.
| |
Collapse
|
11
|
Rani L, Ranjan Sahu M, Chandra Mondal A. Age-related Mitochondrial Dysfunction in Parkinson's Disease: New Insights Into the Disease Pathology. Neuroscience 2022; 499:152-169. [PMID: 35839924 DOI: 10.1016/j.neuroscience.2022.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/13/2022] [Accepted: 07/07/2022] [Indexed: 12/14/2022]
Abstract
Aging is a progressive loss of physiological function that increases risk of disease and death. Among the many factors that contribute to human aging, mitochondrial dysfunction has emerged as one of the most prominent features of the aging process. It has been linked to the development of various age-related pathologies, including Parkinson's disease (PD). Mitochondria has a complex quality control system that ensures mitochondrial integrity and function. Perturbations in these mitochondrial mechanisms have long been linked to various age-related neurological disorders. Even though research has shed light on several aspects of the disease pathology, the underlying mechanism of age-related factors responsible for individuals developing this disease is still unknown. This review article aims to discuss the role of mitochondria in the transition from normal brain aging to pathological brain aging, which leads to the progression of PD. We have discussed the emerging evidence on how age-related disruption of mitochondrial quality control mechanisms contributes to the development of PD-related pathophysiology.
Collapse
Affiliation(s)
- Linchi Rani
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
| | - Manas Ranjan Sahu
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India.
| |
Collapse
|
12
|
Ridderinkhof KR, Krugers HJ. Horizons in Human Aging Neuroscience: From Normal Neural Aging to Mental (Fr)Agility. Front Hum Neurosci 2022; 16:815759. [PMID: 35845248 PMCID: PMC9277589 DOI: 10.3389/fnhum.2022.815759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
While aging is an important risk factor for neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease, age-related cognitive decline can also manifest without apparent neurodegenerative changes. In this review, we discuss molecular, cellular, and network changes that occur during normal aging in the absence of neurodegenerative disease. Emerging findings reveal that these changes include metabolic alterations, oxidative stress, DNA damage, inflammation, calcium dyshomeostasis, and several other hallmarks of age-related neural changes that do not act on their own, but are often interconnected and together may underlie age-related alterations in brain plasticity and cognitive function. Importantly, age-related cognitive decline may not be reduced to a single neurobiological cause, but should instead be considered in terms of a densely connected system that underlies age-related cognitive alterations. We speculate that a decline in one hallmark of neural aging may trigger a decline in other, otherwise thus far stable subsystems, thereby triggering a cascade that may at some point also incur a decline of cognitive functions and mental well-being. Beyond studying the effects of these factors in isolation, considerable insight may be gained by studying the larger picture that entails a representative collection of such factors and their interactions, ranging from molecules to neural networks. Finally, we discuss some potential interventions that may help to prevent these alterations, thereby reducing cognitive decline and mental fragility, and enhancing mental well-being, and healthy aging.
Collapse
Affiliation(s)
- K. Richard Ridderinkhof
- Department of Psychology, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Center for Brain and Cognition (ABC), University of Amsterdam, Amsterdam, Netherlands
| | - Harm J. Krugers
- Amsterdam Center for Brain and Cognition (ABC), University of Amsterdam, Amsterdam, Netherlands
- SILS-CNS, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
13
|
Mokrane N, Snabi Y, Cens T, Guiramand J, Charnet P, Bertaud A, Menard C, Rousset M, de Jesus Ferreira MC, Thibaud JB, Cohen-Solal C, Vignes M, Roussel J. Manipulations of Glutathione Metabolism Modulate IP 3-Mediated Store-Operated Ca 2+ Entry on Astroglioma Cell Line. Front Aging Neurosci 2022; 13:785727. [PMID: 34975458 PMCID: PMC8719003 DOI: 10.3389/fnagi.2021.785727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/01/2021] [Indexed: 02/03/2023] Open
Abstract
The regulation of the redox status involves the activation of intracellular pathways as Nrf2 which provides hormetic adaptations against oxidative stress in response to environmental stimuli. In the brain, Nrf2 activation upregulates the formation of glutathione (GSH) which is the primary antioxidant system mainly produced by astrocytes. Astrocytes have also been shown to be themselves the target of oxidative stress. However, how changes in the redox status itself could impact the intracellular Ca2+ homeostasis in astrocytes is not known, although this could be of great help to understand the neuronal damage caused by oxidative stress. Indeed, intracellular Ca2+ changes in astrocytes are crucial for their regulatory actions on neuronal networks. We have manipulated GSH concentration in astroglioma cells with selective inhibitors and activators of the enzymes involved in the GSH cycle and analyzed how this could modify Ca2+ homeostasis. IP3-mediated store-operated calcium entry (SOCE), obtained after store depletion elicited by Gq-linked purinergic P2Y receptors activation, are either sensitized or desensitized, following GSH depletion or increase, respectively. The desensitization may involve decreased expression of the proteins STIM2, Orai1, and Orai3 which support SOCE mechanism. The sensitization process revealed by exposing cells to oxidative stress likely involves the increase in the activity of Calcium Release-Activated Channels (CRAC) and/or in their membrane expression. In addition, we observe that GSH depletion drastically impacts P2Y receptor-mediated changes in membrane currents, as evidenced by large increases in Ca2+-dependent K+ currents. We conclude that changes in the redox status of astrocytes could dramatically modify Ca2+ responses to Gq-linked GPCR activation in both directions, by impacting store-dependent Ca2+-channels, and thus modify cellular excitability under purinergic stimulation.
Collapse
Affiliation(s)
- Nawfel Mokrane
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Yassin Snabi
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Thierry Cens
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France
| | - Janique Guiramand
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France
| | - Pierre Charnet
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France
| | - Anaïs Bertaud
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Claudine Menard
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Matthieu Rousset
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France
| | - Marie-Céleste de Jesus Ferreira
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | | | - Catherine Cohen-Solal
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Michel Vignes
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Julien Roussel
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| |
Collapse
|
14
|
Hastings N, Kuan WL, Osborne A, Kotter MRN. Therapeutic Potential of Astrocyte Transplantation. Cell Transplant 2022; 31:9636897221105499. [PMID: 35770772 PMCID: PMC9251977 DOI: 10.1177/09636897221105499] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell transplantation is an attractive treatment strategy for a variety of brain disorders, as it promises to replenish lost functions and rejuvenate the brain. In particular, transplantation of astrocytes has come into light recently as a therapy for amyotrophic lateral sclerosis (ALS); moreover, grafting of astrocytes also showed positive results in models of other conditions ranging from neurodegenerative diseases of older age to traumatic injury and stroke. Despite clear differences in etiology, disorders such as ALS, Parkinson's, Alzheimer's, and Huntington's diseases, as well as traumatic injury and stroke, converge on a number of underlying astrocytic abnormalities, which include inflammatory changes, mitochondrial damage, calcium signaling disturbance, hemichannel opening, and loss of glutamate transporters. In this review, we examine these convergent pathways leading to astrocyte dysfunction, and explore the existing evidence for a therapeutic potential of transplantation of healthy astrocytes in various models. Existing literature presents a wide variety of methods to generate astrocytes, or relevant precursor cells, for subsequent transplantation, while described outcomes of this type of treatment also differ between studies. We take technical differences between methodologies into account to understand the variability of therapeutic benefits, or lack thereof, at a deeper level. We conclude by discussing some key requirements of an astrocyte graft that would be most suitable for clinical applications.
Collapse
Affiliation(s)
- Nataly Hastings
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Wei-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Andrew Osborne
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Mark R N Kotter
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
15
|
Wang QQ, Yin G, Huang JR, Xi SJ, Qian F, Lee RX, Peng XC, Tang FR. Ionizing Radiation-Induced Brain Cell Aging and the Potential Underlying Molecular Mechanisms. Cells 2021; 10:3570. [PMID: 34944078 PMCID: PMC8700624 DOI: 10.3390/cells10123570] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 01/10/2023] Open
Abstract
Population aging is occurring rapidly worldwide, challenging the global economy and healthcare services. Brain aging is a significant contributor to various age-related neurological and neuropsychological disorders, including Alzheimer's disease and Parkinson's disease. Several extrinsic factors, such as exposure to ionizing radiation, can accelerate senescence. Multiple human and animal studies have reported that exposure to ionizing radiation can have varied effects on organ aging and lead to the prolongation or shortening of life span depending on the radiation dose or dose rate. This paper reviews the effects of radiation on the aging of different types of brain cells, including neurons, microglia, astrocytes, and cerebral endothelial cells. Further, the relevant molecular mechanisms are discussed. Overall, this review highlights how radiation-induced senescence in different cell types may lead to brain aging, which could result in the development of various neurological and neuropsychological disorders. Therefore, treatment targeting radiation-induced oxidative stress and neuroinflammation may prevent radiation-induced brain aging and the neurological and neuropsychological disorders it may cause.
Collapse
Affiliation(s)
- Qin-Qi Wang
- Laboratory of Oncology, Center for Molecular Medicine, Health Science Center, School of Basic Medicine, Yangtze University, Jingzhou 434023, China; (Q.-Q.W.); (S.-J.X.)
- Health Science Center, Department of Pathophysiology, School of Basic Medicine, Yangtze University, Jingzhou 434023, China
| | - Gang Yin
- Department of Neurology, Jingzhou Central Hospital, Jingzhou 434023, China;
| | - Jiang-Rong Huang
- Health Science Center, Department of Integrative Medicine, School of Health Sciences, Yangtze University, Jingzhou 434023, China;
| | - Shi-Jun Xi
- Laboratory of Oncology, Center for Molecular Medicine, Health Science Center, School of Basic Medicine, Yangtze University, Jingzhou 434023, China; (Q.-Q.W.); (S.-J.X.)
- Health Science Center, Department of Pathophysiology, School of Basic Medicine, Yangtze University, Jingzhou 434023, China
| | - Feng Qian
- Health Science Center, Department of Physiology, School of Basic Medicine, Yangtze University, Jingzhou 434023, China;
| | - Rui-Xue Lee
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore;
| | - Xiao-Chun Peng
- Laboratory of Oncology, Center for Molecular Medicine, Health Science Center, School of Basic Medicine, Yangtze University, Jingzhou 434023, China; (Q.-Q.W.); (S.-J.X.)
- Health Science Center, Department of Pathophysiology, School of Basic Medicine, Yangtze University, Jingzhou 434023, China
| | - Feng-Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore;
| |
Collapse
|
16
|
Sutherland TC, Sefiani A, Horvat D, Huntington TE, Lei Y, West AP, Geoffroy CG. Age-Dependent Decline in Neuron Growth Potential and Mitochondria Functions in Cortical Neurons. Cells 2021; 10:1625. [PMID: 34209640 PMCID: PMC8306398 DOI: 10.3390/cells10071625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022] Open
Abstract
The age of incidence of spinal cord injury (SCI) and the average age of people living with SCI is continuously increasing. However, SCI is extensively modeled in young adult animals, hampering translation of research to clinical applications. While there has been significant progress in manipulating axon growth after injury, the impact of aging is still unknown. Mitochondria are essential to successful neurite and axon growth, while aging is associated with a decline in mitochondrial functions. Using isolation and culture of adult cortical neurons, we analyzed mitochondrial changes in 2-, 6-, 12- and 18-month-old mice. We observed reduced neurite growth in older neurons. Older neurons also showed dysfunctional respiration, reduced membrane potential, and altered mitochondrial membrane transport proteins; however, mitochondrial DNA (mtDNA) abundance and cellular ATP were increased. Taken together, these data suggest that dysfunctional mitochondria in older neurons may be associated with the age-dependent reduction in neurite growth. Both normal aging and traumatic injury are associated with mitochondrial dysfunction, posing a challenge for an aging SCI population as the two elements can combine to worsen injury outcomes. The results of this study highlight this as an area of great interest in CNS trauma.
Collapse
Affiliation(s)
- Theresa C. Sutherland
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX 77807, USA; (T.C.S.); (A.S.); (D.H.); (T.E.H.)
| | - Arthur Sefiani
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX 77807, USA; (T.C.S.); (A.S.); (D.H.); (T.E.H.)
| | - Darijana Horvat
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX 77807, USA; (T.C.S.); (A.S.); (D.H.); (T.E.H.)
| | - Taylor E. Huntington
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX 77807, USA; (T.C.S.); (A.S.); (D.H.); (T.E.H.)
| | - Yuanjiu Lei
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807, USA; (Y.L.); (A.P.W.)
| | - A. Phillip West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807, USA; (Y.L.); (A.P.W.)
| | - Cédric G. Geoffroy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX 77807, USA; (T.C.S.); (A.S.); (D.H.); (T.E.H.)
| |
Collapse
|
17
|
Zia A, Pourbagher-Shahri AM, Farkhondeh T, Samarghandian S. Molecular and cellular pathways contributing to brain aging. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2021; 17:6. [PMID: 34118939 PMCID: PMC8199306 DOI: 10.1186/s12993-021-00179-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022]
Abstract
Aging is the leading risk factor for several age-associated diseases such as neurodegenerative diseases. Understanding the biology of aging mechanisms is essential to the pursuit of brain health. In this regard, brain aging is defined by a gradual decrease in neurophysiological functions, impaired adaptive neuroplasticity, dysregulation of neuronal Ca2+ homeostasis, neuroinflammation, and oxidatively modified molecules and organelles. Numerous pathways lead to brain aging, including increased oxidative stress, inflammation, disturbances in energy metabolism such as deregulated autophagy, mitochondrial dysfunction, and IGF-1, mTOR, ROS, AMPK, SIRTs, and p53 as central modulators of the metabolic control, connecting aging to the pathways, which lead to neurodegenerative disorders. Also, calorie restriction (CR), physical exercise, and mental activities can extend lifespan and increase nervous system resistance to age-associated neurodegenerative diseases. The neuroprotective effect of CR involves increased protection against ROS generation, maintenance of cellular Ca2+ homeostasis, and inhibition of apoptosis. The recent evidence about the modem molecular and cellular methods in neurobiology to brain aging is exhibiting a significant potential in brain cells for adaptation to aging and resistance to neurodegenerative disorders.
Collapse
Affiliation(s)
- Aliabbas Zia
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ali Mohammad Pourbagher-Shahri
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), 9717853577 Birjand, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
18
|
Melzer TM, Manosso LM, Yau SY, Gil-Mohapel J, Brocardo PS. In Pursuit of Healthy Aging: Effects of Nutrition on Brain Function. Int J Mol Sci 2021; 22:5026. [PMID: 34068525 PMCID: PMC8126018 DOI: 10.3390/ijms22095026] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
Consuming a balanced, nutritious diet is important for maintaining health, especially as individuals age. Several studies suggest that consuming a diet rich in antioxidants and anti-inflammatory components such as those found in fruits, nuts, vegetables, and fish may reduce age-related cognitive decline and the risk of developing various neurodegenerative diseases. Numerous studies have been published over the last decade focusing on nutrition and how this impacts health. The main objective of the current article is to review the data linking the role of diet and nutrition with aging and age-related cognitive decline. Specifically, we discuss the roles of micronutrients and macronutrients and provide an overview of how the gut microbiota-gut-brain axis and nutrition impact brain function in general and cognitive processes in particular during aging. We propose that dietary interventions designed to optimize the levels of macro and micronutrients and maximize the functioning of the microbiota-gut-brain axis can be of therapeutic value for improving cognitive functioning, particularly during aging.
Collapse
Affiliation(s)
- Thayza Martins Melzer
- Neuroscience Graduate Program, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil;
| | - Luana Meller Manosso
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma 88806-000, SC, Brazil;
| | - Suk-yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Joana Gil-Mohapel
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada
| | - Patricia S. Brocardo
- Neuroscience Graduate Program, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil;
| |
Collapse
|
19
|
Zhang C, Yan Z, Maknojia A, Riquelme MA, Gu S, Booher G, Wallace DJ, Bartanusz V, Goswami A, Xiong W, Zhang N, Mader MJ, An Z, Sayre NL, Jiang JX. Inhibition of astrocyte hemichannel improves recovery from spinal cord injury. JCI Insight 2021; 6:134611. [PMID: 33682795 PMCID: PMC8021110 DOI: 10.1172/jci.insight.134611] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/27/2021] [Indexed: 11/25/2022] Open
Abstract
Spinal cord injury (SCI) causes severe disability, and the current inability to restore function to the damaged spinal cord leads to lasting detrimental consequences to patients. One strategy to reduce SCI morbidity involves limiting the spread of secondary damage after injury. Previous studies have shown that connexin 43 (Cx43), a gap junction protein richly expressed in spinal cord astrocytes, is a potential mediator of secondary damage. Here, we developed a specific inhibitory antibody, mouse-human chimeric MHC1 antibody (MHC1), that inhibited Cx43 hemichannels, but not gap junctions, and reduced secondary damage in 2 incomplete SCI mouse models. MHC1 inhibited the activation of Cx43 hemichannels in both primary spinal astrocytes and astrocytes in situ. In both SCI mouse models, administration of MHC1 after SCI significantly improved hind limb locomotion function. Remarkably, a single administration of MHC1 30 minutes after injury improved the recovery up to 8 weeks post-SCI. Moreover, MHC1 treatment decreased gliosis and lesion sizes, increased white and gray matter sparing, and improved neuronal survival. Together, these results suggest that inhibition of Cx43 hemichannel function after traumatic SCI reduces secondary damage, limits perilesional gliosis, and improves functional recovery. By targeting hemichannels specifically with an antibody, this study provides a potentially new, innovative therapeutic approach in treating SCI.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhao Yan
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - Asif Maknojia
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Manuel A Riquelme
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Grant Booher
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - David J Wallace
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Viktor Bartanusz
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Akshay Goswami
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Michael J Mader
- Audie L. Murphy VA Hospital, South Texas Veteran's Health Care System, San Antonio, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Naomi L Sayre
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Audie L. Murphy VA Hospital, South Texas Veteran's Health Care System, San Antonio, Texas, USA
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
20
|
Dorigatti AO, Hussong SA, Hernandez SF, Sills AM, Salmon AB, Galvan V. Primary neuron and astrocyte cultures from postnatal Callithrix jacchus: a non-human primate in vitro model for research in neuroscience, nervous system aging, and neurological diseases of aging. GeroScience 2021; 43:115-124. [PMID: 33063253 PMCID: PMC8050148 DOI: 10.1007/s11357-020-00284-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 10/08/2020] [Indexed: 01/05/2023] Open
Abstract
The ability to generate in vitro cultures of neuronal cells has been instrumental in advancing our understanding of the nervous system. Rodent models have been the principal source of brain cells used in primary cultures for over a century, providing insights that are widely applicable to human diseases. However, therapeutic agents that showed benefit in rodent models, particularly those pertaining to aging and age-associated dementias, have frequently failed in clinical trials. This discrepancy established a potential "translational gap" between human and rodent studies that may at least partially be explained by the phylogenetic distance between rodent and primate species. Several non-human primate (NHP) species, including the common marmoset (Callithrix jacchus), have been used extensively in neuroscience research, but in contrast to rodent models, practical approaches to the generation of primary cell culture systems amenable to molecular studies that can inform in vivo studies are lacking. Marmosets are a powerful model in biomedical research and particularly in studies of aging and age-associated diseases because they exhibit an aging phenotype similar to humans. Here, we report a practical method to culture primary marmoset neurons and astrocytes from brains of medically euthanized postnatal day 0 (P0) marmoset newborns that yield highly pure primary neuron and astrocyte cultures. Primary marmoset neuron and astrocyte cultures can be generated reliably to provide a powerful NHP in vitro model in neuroscience research that may enable mechanistic studies of nervous system aging and of age-related neurodegenerative disorders. Because neuron and astrocyte cultures can be used in combination with in vivo approaches in marmosets, primary marmoset neuron and astrocyte cultures may help bridge the current translational gap between basic and clinical studies in nervous system aging and age-associated neurological diseases.
Collapse
Affiliation(s)
- Angela O Dorigatti
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, 15355 Lambda Drive, STCBM 3.200.8, San Antonio, TX, 78245, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Stacy A Hussong
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, 15355 Lambda Drive, STCBM 3.200.8, San Antonio, TX, 78245, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Stephen F Hernandez
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, 15355 Lambda Drive, STCBM 3.200.8, San Antonio, TX, 78245, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Aubrey M Sills
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Adam B Salmon
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- South Texas Veterans Health Care System, San Antonio, TX, USA
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Veronica Galvan
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, 15355 Lambda Drive, STCBM 3.200.8, San Antonio, TX, 78245, USA.
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- South Texas Veterans Health Care System, San Antonio, TX, USA.
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
21
|
Oksanen M, Hyötyläinen I, Trontti K, Rolova T, Wojciechowski S, Koskuvi M, Viitanen M, Levonen A, Hovatta I, Roybon L, Lehtonen Š, Kanninen KM, Hämäläinen RH, Koistinaho J. NF-E2-related factor 2 activation boosts antioxidant defenses and ameliorates inflammatory and amyloid properties in human Presenilin-1 mutated Alzheimer's disease astrocytes. Glia 2020; 68:589-599. [PMID: 31670864 PMCID: PMC7003860 DOI: 10.1002/glia.23741] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/11/2019] [Accepted: 10/11/2019] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a common dementia affecting a vast number of individuals and significantly impairing quality of life. Despite extensive research in animal models and numerous promising treatment trials, there is still no curative treatment for AD. Astrocytes, the most common cell type of the central nervous system, have been shown to play a role in the major AD pathologies, including accumulation of amyloid plaques, neuroinflammation, and oxidative stress. Here, we show that inflammatory stimulation leads to metabolic activation of human astrocytes and reduces amyloid secretion. On the other hand, the activation of oxidative metabolism leads to increased reactive oxygen species production especially in AD astrocytes. While healthy astrocytes increase glutathione (GSH) release to protect the cells, Presenilin-1-mutated AD patient astrocytes do not. Thus, chronic inflammation is likely to induce oxidative damage in AD astrocytes. Activation of NRF2, the major regulator of cellular antioxidant defenses, encoded by the NFE2L2 gene, poses several beneficial effects on AD astrocytes. We report here that the activation of NRF2 pathway reduces amyloid secretion, normalizes cytokine release, and increases GSH secretion in AD astrocytes. NRF2 induction also activates the metabolism of astrocytes and increases the utilization of glycolysis. Taken together, targeting NRF2 in astrocytes could be a potent therapeutic strategy in AD.
Collapse
Affiliation(s)
- Minna Oksanen
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Ida Hyötyläinen
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Kalevi Trontti
- Neuroscience CenterUniversity of HelsinkiHelsinkiFinland
- SleepWell Research Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Psychology and LogopedicsUniversity of HelsinkiHelsinkiFinland
| | - Taisia Rolova
- Neuroscience CenterUniversity of HelsinkiHelsinkiFinland
| | - Sara Wojciechowski
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Marja Koskuvi
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
- Neuroscience CenterUniversity of HelsinkiHelsinkiFinland
| | - Matti Viitanen
- Department of GeriatricsUniversity of Turku, Turku City HospitalTurkuFinland
- Department of GeriatricsKarolinska Institutet and Karolinska University HospitalStockholmSweden
| | - Anna‐Liisa Levonen
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Iiris Hovatta
- Neuroscience CenterUniversity of HelsinkiHelsinkiFinland
- SleepWell Research Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Psychology and LogopedicsUniversity of HelsinkiHelsinkiFinland
| | - Laurent Roybon
- Department of Experimental Medical Science and MultiPark and Lund Stem Cell CenterFaculty of Medicine, Lund UniversityLundSweden
| | - Šárka Lehtonen
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
- Neuroscience CenterUniversity of HelsinkiHelsinkiFinland
| | - Katja M. Kanninen
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Riikka H. Hämäläinen
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Jari Koistinaho
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
- Neuroscience CenterUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
22
|
Abstract
Aging brain becomes susceptible to neurodegenerative diseases due to the shifting of microglia and astrocyte phenotypes to an active “pro-inflammatory” state, causing chronic low-grade neuroinflammation. Despite the fact that the role of neuroinflammation during aging has been extensively studied in recent years, the underlying causes remain unclear. The identification of relevant proteins and understanding their potential roles in neuroinflammation can help explain their potential of becoming biomarkers in the aging brain and as drug targets for prevention and treatment. This will eventually reduce the chances of developing neurodegenerative diseases and promote healthier lives in the elderly. In this review, we have summarized the morphological and cellular changes in the aging brain, the effects of age-related neuroinflammation, and the potential role of cofilin-1 during neuroinflammation. We also discuss other factors contributing to brain aging and neuroinflammation.
Collapse
Affiliation(s)
- Amsha S Alsegiani
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| |
Collapse
|
23
|
Abdelkarim D, Zhao Y, Turner MP, Sivakolundu DK, Lu H, Rypma B. A neural-vascular complex of age-related changes in the human brain: Anatomy, physiology, and implications for neurocognitive aging. Neurosci Biobehav Rev 2019; 107:927-944. [DOI: 10.1016/j.neubiorev.2019.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 08/02/2019] [Accepted: 09/02/2019] [Indexed: 01/09/2023]
|
24
|
Telomerase Reverse Transcriptase and p53 Regulate Mammalian Peripheral Nervous System and CNS Axon Regeneration Downstream of c-Myc. J Neurosci 2019; 39:9107-9118. [PMID: 31597725 DOI: 10.1523/jneurosci.0419-19.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 09/04/2019] [Accepted: 09/29/2019] [Indexed: 12/12/2022] Open
Abstract
Although several genes have been identified to promote axon regeneration in the CNS, our understanding of the molecular mechanisms by which mammalian axon regeneration is regulated is still limited and fragmented. Here by using female mouse sensory axon and optic nerve regeneration as model systems, we reveal an unexpected role of telomerase reverse transcriptase (TERT) in regulation of axon regeneration. We also provide evidence that TERT and p53 act downstream of c-Myc to control sensory axon regeneration. More importantly, overexpression of p53 in sensory neurons and retinal ganglion cells is sufficient to promote sensory axon and optic never regeneration, respectively. The study reveals a novel c-Myc-TERT-p53 signaling pathway, expanding horizons for novel approaches promoting CNS axon regeneration.SIGNIFICANCE STATEMENT Despite significant progress during the past decade, our understanding of the molecular mechanisms by which mammalian CNS axon regeneration is regulated is still fragmented. By using sensory axon and optic nerve regeneration as model systems, the study revealed an unexpected role of telomerase reverse transcriptase (TERT) in regulation of axon regeneration. The results also delineated a c-Myc-TERT-p53 pathway in controlling axon growth. Last, our results demonstrated that p53 alone was sufficient to promote sensory axon and optic nerve regeneration in vivo Collectively, the study not only revealed a new mechanisms underlying mammalian axon regeneration, but also expanded the pool of potential targets that can be manipulated to enhance CNS axon regeneration.
Collapse
|
25
|
Oksanen M, Lehtonen S, Jaronen M, Goldsteins G, Hämäläinen RH, Koistinaho J. Astrocyte alterations in neurodegenerative pathologies and their modeling in human induced pluripotent stem cell platforms. Cell Mol Life Sci 2019; 76:2739-2760. [PMID: 31016348 PMCID: PMC6588647 DOI: 10.1007/s00018-019-03111-7] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 04/06/2019] [Accepted: 04/16/2019] [Indexed: 12/12/2022]
Abstract
Astrocytes are the most abundant cell type in the brain. They were long considered only as passive support for neuronal cells. However, recent data have revealed many active roles for these cells both in maintenance of the normal physiological homeostasis in the brain as well as in neurodegeneration and disease. Moreover, human astrocytes have been found to be much more complex than their rodent counterparts, and to date, astrocytes are known to actively participate in a multitude of processes such as neurotransmitter uptake and recycling, gliotransmitter release, neuroenergetics, inflammation, modulation of synaptic activity, ionic balance, maintenance of the blood-brain barrier, and many other crucial functions of the brain. This review focuses on the role of astrocytes in human neurodegenerative disease and the potential of the novel stem cell-based platforms in modeling astrocytic functions in health and in disease.
Collapse
Affiliation(s)
- Minna Oksanen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Sarka Lehtonen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, PO. Box 63, 00290, Helsinki, Finland
| | - Merja Jaronen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Gundars Goldsteins
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Riikka H Hämäläinen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Jari Koistinaho
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland.
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, PO. Box 63, 00290, Helsinki, Finland.
| |
Collapse
|
26
|
Alarcón-Aguilar A, Luna-López A, López-Diazguerrero NE, Königsberg M. The Effect of MPP+ on the Viability of Primary Cortical Astrocytes Isolated from Female and Male Wistar Rats of Different Ages. Cell Mol Neurobiol 2019; 39:321-328. [PMID: 30539419 PMCID: PMC11469902 DOI: 10.1007/s10571-018-0643-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023]
Abstract
Although age is known to be the main risk for developing chronic and neurodegenerative diseases, those illnesses have a different prevalence depending on the sex. It has been questioned whether genetic and hormonal differences are preserved in primary cultures from individuals of different genders. Therefore, here we studied the susceptibility of astrocytes, obtained from female and male Wistar rats of different ages (newborn, 9 and 24 months-old), to the well-known toxin MPP+ after 2 weeks in vitro, at different concentrations and exposure times. Our results showed that there are no variances due to gender, but that there are important differences associated to age in terms of the viability against this toxin.
Collapse
Affiliation(s)
- Adriana Alarcón-Aguilar
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, Ciudad de México, Mexico
| | | | | | - Mina Königsberg
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, Ciudad de México, Mexico.
- División de Ciencias Biológicas y de la Salud, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, A.P. 55-535, C.P 09340, Ciudad de México, Mexico.
| |
Collapse
|
27
|
Abstract
By 2050, the aging population is predicted to expand by over 100%. Considering this rapid growth, and the additional strain it will place on healthcare resources because of age-related impairments, it is vital that researchers gain a deeper understanding of the cellular interactions that occur with normal aging. A variety of mammalian cell types have been shown to become compromised with age, each with a unique potential to contribute to disease formation in the aging body. Astrocytes represent the largest group of glial cells and are responsible for a variety of essential functions in the healthy central nervous system (CNS). Like other cell types, aging can cause a loss of normal function in astrocytes which reduces their ability to properly maintain a healthy CNS environment, negatively alters their interactions with neighboring cells, and contribute to the heightened inflammatory state characteristic of aging. The goal of this review article is to consolidate the knowledge and research to date regarding the role of astrocytes in aging. In specific, this review article will focus on the morphology and molecular profile of aged astrocytes, the consequence of astrocyte dysfunction on homeostatic functions during aging, and the role of astrocytes in age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Alexandra L Palmer
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Shalina S Ousman
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Departments of Clinical Neurosciences and Cell Biology & Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
28
|
Mattson MP, Arumugam TV. Hallmarks of Brain Aging: Adaptive and Pathological Modification by Metabolic States. Cell Metab 2018; 27:1176-1199. [PMID: 29874566 PMCID: PMC6039826 DOI: 10.1016/j.cmet.2018.05.011] [Citation(s) in RCA: 723] [Impact Index Per Article: 103.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/02/2018] [Accepted: 05/15/2018] [Indexed: 02/06/2023]
Abstract
During aging, the cellular milieu of the brain exhibits tell-tale signs of compromised bioenergetics, impaired adaptive neuroplasticity and resilience, aberrant neuronal network activity, dysregulation of neuronal Ca2+ homeostasis, the accrual of oxidatively modified molecules and organelles, and inflammation. These alterations render the aging brain vulnerable to Alzheimer's and Parkinson's diseases and stroke. Emerging findings are revealing mechanisms by which sedentary overindulgent lifestyles accelerate brain aging, whereas lifestyles that include intermittent bioenergetic challenges (exercise, fasting, and intellectual challenges) foster healthy brain aging. Here we provide an overview of the cellular and molecular biology of brain aging, how those processes interface with disease-specific neurodegenerative pathways, and how metabolic states influence brain health.
Collapse
Affiliation(s)
- Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
29
|
Kawano H, Oyabu K, Yamamoto H, Eto K, Adaniya Y, Kubota K, Watanabe T, Hirano-Iwata A, Nabekura J, Katsurabayashi S, Iwasaki K. Astrocytes with previous chronic exposure to amyloid β-peptide fragment 1-40 suppress excitatory synaptic transmission. J Neurochem 2017; 143:624-634. [PMID: 29076533 DOI: 10.1111/jnc.14247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/27/2017] [Accepted: 10/13/2017] [Indexed: 11/28/2022]
Abstract
Synaptic dysfunction and neuronal death are responsible for cognitive and behavioral deficits in Alzheimer's disease (AD). It is well known that such neurological abnormalities are preceded by long-term exposure of amyloid β-peptide (Aβ) and/or hyperphosphorylated tau prior. In addition to the neurological deficit, astrocytes as a major glial cell type in the brain, significantly participate in the neuropathogenic mechanisms underlying synaptic modulation. Although astrocytes play a significant key role in modulating synaptic transmission, little is known on whether astrocyte dysfunction caused by such long-term Aβ exposure affects synapse formation and function. Here, we show that synapse formation and synaptic transmission are attenuated in hippocampal-naïve neurons co-cultured with astrocytes that have previously experienced chronic Aβ1-40 exposure. In this abnormal astrocytic condition, hippocampal neurons exhibit decrements of evoked excitatory post-synaptic currents (EPSCs) and miniature EPSC frequency. Furthermore, size of readily releasable synaptic pools and number of excitatory synapses were also significantly decreased. Contrary to these negative effects, release probability at individual synapses was significantly increased in the same astrocytic condition. Taken together, our data indicate that lower synaptic transmission caused by astrocytes previously, and chronically, exposed to Aβ1-40 is attributable to a small number of synapses with higher release probability.
Collapse
Affiliation(s)
- Hiroyuki Kawano
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kohei Oyabu
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Hideaki Yamamoto
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aoba-ku, Sendai, Japan
| | - Kei Eto
- Division of Homeostatic Development, Department of Fundamental Neuroscience, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Yuna Adaniya
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kaori Kubota
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.,A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, Japan
| | - Takuya Watanabe
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.,A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, Japan
| | - Ayumi Hirano-Iwata
- Advanced Institute for Materials Research, Tohoku University, Aoba-ku, Sendai, Japan.,Research Institute of Electrical Communication, Tohoku University, Aoba-ku, Sendai, Japan
| | - Junichi Nabekura
- Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan.,Division of Homeostatic Development, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Japan.,CREST, Japan Science and Technology Agency (JST), Kawaguchi, Japan
| | - Shutaro Katsurabayashi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Katsunori Iwasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.,A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
30
|
Sayre NL, Sifuentes M, Holstein D, Cheng SY, Zhu X, Lechleiter JD. Stimulation of astrocyte fatty acid oxidation by thyroid hormone is protective against ischemic stroke-induced damage. J Cereb Blood Flow Metab 2017; 37:514-527. [PMID: 26873887 PMCID: PMC5381439 DOI: 10.1177/0271678x16629153] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We previously demonstrated that stimulation of astrocyte mitochondrial ATP production via P2Y1 receptor agonists was neuroprotective after cerebral ischemic stroke. Another mechanism that increases ATP production is fatty acid oxidation (FAO). We show that in primary human astrocytes, FAO and ATP production are stimulated by 3,3,5 triiodo-l-thyronine (T3). We tested whether T3-stimulated FAO enhances neuroprotection, and show that T3 increased astrocyte survival after either hydrogen peroxide exposure or oxygen glucose deprivation. T3-mediated ATP production and protection were both eliminated with etomoxir, an inhibitor of FAO. T3-mediated protection in vitro was also dependent on astrocytes expressing HADHA (hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase), which we previously showed was critical for T3-mediated FAO in fibroblasts. Consistent with previous reports, T3-treatment decreased stroke volumes in mice. While T3 decreased stroke volume in etomoxir-treated mice, T3 had no protective effect on stroke volume in HADHA +/- mice or in mice unable to upregulate astrocyte-specific energy production. In vivo, 95% of HADHA co-localize with glial-fibrillary acidic protein, suggesting the effect of HADHA is astrocyte mediated. These results suggest that astrocyte-FAO modulates lesion size and is required for T3-mediated neuroprotection post-stroke. To our knowledge, this is the first report of a neuroprotective role for FAO in the brain.
Collapse
Affiliation(s)
- Naomi L Sayre
- 1 Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mikaela Sifuentes
- 2 Department of Cellular & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Deborah Holstein
- 2 Department of Cellular & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sheue-Yann Cheng
- 3 Gene Regulation Section, Laboratory of Molecular Biology, National Institutes of Health, Bethesda, MD, 20892
| | - Xuguang Zhu
- 3 Gene Regulation Section, Laboratory of Molecular Biology, National Institutes of Health, Bethesda, MD, 20892
| | - James D Lechleiter
- 2 Department of Cellular & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,4 Department of Cellular & Structural Biology, South Texas Research Facility Neuroscience Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
31
|
Li X, Floriddia EM, Toskas K, Fernandes KJL, Guérout N, Barnabé-Heider F. Regenerative Potential of Ependymal Cells for Spinal Cord Injuries Over Time. EBioMedicine 2016; 13:55-65. [PMID: 27818039 PMCID: PMC5264475 DOI: 10.1016/j.ebiom.2016.10.035] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/24/2016] [Accepted: 10/24/2016] [Indexed: 12/22/2022] Open
Abstract
Stem cells have a high therapeutic potential for the treatment of spinal cord injury (SCI). We have shown previously that endogenous stem cell potential is confined to ependymal cells in the adult spinal cord which could be targeted for non-invasive SCI therapy. However, ependymal cells are an understudied cell population. Taking advantage of transgenic lines, we characterize the appearance and potential of ependymal cells during development. We show that spinal cord stem cell potential in vitro is contained within these cells by birth. Moreover, juvenile cultures generate more neurospheres and more oligodendrocytes than adult ones. Interestingly, juvenile ependymal cells in vivo contribute to glial scar formation after severe but not mild SCI, due to a more effective sealing of the lesion by other glial cells. This study highlights the importance of the age-dependent potential of stem cells and post-SCI environment in order to utilize ependymal cell's regenerative potential.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Elisa M Floriddia
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Karl J L Fernandes
- Department of Neurosciences, Research Center of the University of Montreal Hospital (CRCHUM), QC H2X 0A9 Montreal, Canada
| | - Nicolas Guérout
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden; Normandie Université, UNIROUEN, EA3830-GRHV, 76000 Rouen, France; Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France.
| | | |
Collapse
|
32
|
Wang F, Zhang L. p15(INK4b) regulates cell cycle signaling in hippocampal astrocytes of aged rats. Aging Clin Exp Res 2016; 28:813-21. [PMID: 26526028 DOI: 10.1007/s40520-015-0484-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 10/19/2015] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND AIMS Cyclin-dependent kinase inhibitor p15(INK4b) is thought to be an important player in regulating astrocytic cell cycle. However, little is known with regard to the expression of p15(INK4b) and its function in hippocampal astrocytes. This study evaluated the expression of p15(INK4b) and its function during different development stages in hippocampal astrocytes. METHODS In this study, we cultured hippocampal astrocytes from neonatal adult and aged rats. The expression of p15(INK4b) in neonatal, adult and aged astrocytes was examined. Short interfering RNA (siRNA) was then used to study the functional effects of p15(INK4b) down-regulation during cell cycle regulation. RESULTS We found the expression of p15(INK4b) in hippocampal astrocytes was detectable on postnatal day 7, was expressed at moderate levels in adult mice (9 months old) astrocytes and peaked in aged rat (24 months old) astrocytes. Incubation with siRNA significantly suppressed p15(INK4b) expression at the mRNA and protein levels in astrocytes. Down-regulation of p15(INK4b) increased [(3)H]-thymidine incorporation into DNA and allowed cells to pass the G0/G1-S checkpoint in aged but not in neonatal or adult astrocytes. CONCLUSIONS These observations suggest p15(INK4b) is expressed at a steady level in neonatal and adult rat hippocampal astrocytes with no effect on cell cycle regulation. Importantly, aged astrocyte cell cycle regulation was significantly affected by high expression levels of p15(INK4b) suggesting a role for p15(INK4b) in cell cycle regulation when it is expressed at high but not moderate or low levels in hippocampal astrocytes.
Collapse
Affiliation(s)
- Fang Wang
- Department of Neurology, The Central Hospital of Wuhan, 26 Shengli Street, Wuhan, 430014, China.
| | - Linhong Zhang
- Department of Neurology, The Central Hospital of Wuhan, 26 Shengli Street, Wuhan, 430014, China
| |
Collapse
|
33
|
Chen Y, Holstein DM, Aime S, Bollo M, Lechleiter JD. Calcineurin β protects brain after injury by activating the unfolded protein response. Neurobiol Dis 2016; 94:139-56. [PMID: 27334877 PMCID: PMC4983525 DOI: 10.1016/j.nbd.2016.06.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/15/2016] [Accepted: 06/17/2016] [Indexed: 11/26/2022] Open
Abstract
The Ca2+-dependent phosphatase, calcineurin (CN) is thought to play a detrimental role in damaged neurons; however, its role in astrocytes is unclear. In cultured astrocytes, CNβ expression increased after treatment with a sarco/endoplasmic reticulum Ca2+-ATPase inhibitor, thapsigargin, and with oxygen and glucose deprivation, an in vitro model of ischemia. Similarly, CNβ was induced in astrocytes in vivo in two different mouse models of brain injury - photothrombotic stroke and traumatic brain injury (TBI). Immunoprecipitation and chemical activation dimerization methods pointed to physical interaction of CNβ with the unfolded protein response (UPR) sensor, protein kinase RNA-like endoplasmic reticulum kinase (PERK). In accordance, induction of CNβ resulted in oligomerization and activation of PERK. Strikingly, the presence of a phosphatase inhibitor did not interfere with CNβ-mediated activation of PERK, suggesting a hitherto undiscovered non-enzymatic role for CNβ. Importantly, the cytoprotective function of CNβ was PERK-dependent both in vitro and in vivo. Loss of CNβ in vivo resulted in a significant increase in cerebral damage, and correlated with a decrease in astrocyte size, PERK activity and glial fibrillary acidic protein (GFAP) expression. Taken together, these data reveal a critical role for the CNβ-PERK axis in not only prolonging astrocyte cell survival but also in modulating astrogliosis after brain injury.
Collapse
Affiliation(s)
- Yanan Chen
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, TX, USA
| | - Deborah M Holstein
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, TX, USA
| | - Sofia Aime
- Instituto de Investigación Médica M y M Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mariana Bollo
- Instituto de Investigación Médica M y M Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - James D Lechleiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, TX, USA; Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, TX, USA.
| |
Collapse
|
34
|
1,3-Dinitrobenzene neurotoxicity - Passage effect in immortalized astrocytes. Neurotoxicology 2016; 53:74-84. [PMID: 26769196 DOI: 10.1016/j.neuro.2015.11.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 11/21/2022]
Abstract
Age-related disturbances in astrocytic mitochondrial function are linked to loss of neuroprotection and decrements in neurological function. The immortalized rat neocortical astrocyte-derived cell line, DI-TNC1, provides a convenient model for the examination of cellular aging processes that are difficult to study in primary cell isolates from aged brain. Successive passages in culture may serve as a surrogate of aging in which time-dependent adaptation to culture conditions may result in altered responses to xenobiotic challenge. To investigate the hypothesis that astrocytic mitochondrial homeostatic function is decreased with time in culture, low passage DI-TNC1 astrocytes (LP; #2-8) and high passage DI-TNC1 astrocytes (HP; #17-28) were exposed to the mitochondrial neurotoxicant 1,3-dinitrobenzene (DNB). Cells were exposed in either monoculture or in co-culture with primary cortical neurons. Astrocyte mitochondrial membrane potential, morphology, ATP production and proliferation were monitored in monoculture, and the ability of DI-TNC1 cells to buffer K(+)-induced neuronal depolarization was examined in co-cultures. In HP DI-TNC1 cells, DNB exposure decreased proliferation, reduced mitochondrial membrane potential and significantly decreased mitochondrial form factor. Low passage DI-TNC1 cells effectively attenuated K(+)-induced neuronal depolarization in the presence of DNB whereas HP counterparts were unable to buffer K(+) in DNB challenge. Following DNB challenge, LP DI-TNC1 cells exhibited greater viability in co-culture than HP. The data provide compelling evidence that there is an abrupt phenotypic change in DI-TNC1 cells between passage #9-16 that significantly diminishes the ability of DI-TNC1 cells to compensate for neurotoxic challenge and provide neuroprotective spatial buffering. Whether or not these functional changes have an in vivo analog in aging brain remains to be determined.
Collapse
|
35
|
Kubik LL, Philbert MA. The role of astrocyte mitochondria in differential regional susceptibility to environmental neurotoxicants: tools for understanding neurodegeneration. Toxicol Sci 2015; 144:7-16. [PMID: 25740792 DOI: 10.1093/toxsci/kfu254] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In recent decades, there has been a significant expansion in our understanding of the role of astrocytes in neuroprotection, including spatial buffering of extracellular ions, secretion of metabolic coenzymes, and synaptic regulation. Astrocytic neuroprotective functions require energy, and therefore require a network of functional mitochondria. Disturbances to astrocytic mitochondrial homeostasis and their ability to produce ATP can negatively impact neural function. Perturbations in astrocyte mitochondrial function may accrue as the result of physiological aging processes or as a consequence of neurotoxicant exposure. Hydrophobic environmental neurotoxicants, such as 1,3-dinitrobenzene and α-chlorohydrin, cause regionally specific spongiform lesions mimicking energy deprivation syndromes. Astrocyte involvement includes mitochondrial damage that either precedes or is accompanied by neuronal damage. Similarly, environmental neurotoxicants that are implicated in the etiology of age-related neurodegenerative conditions cause regionally specific damage in the brain. Based on the regioselective nature of age-related neurodegenerative lesions, chemically induced models of regioselective lesions targeting astrocyte mitochondria can provide insight into age-related susceptibilities in astrocyte mitochondria. Most of the available research to date focuses on neuronal damage in cases of age-related neurodegeneration; however, there is a body of evidence that supports a central mechanistic role for astrocyte mitochondria in the expression of neural injury. Regional susceptibility to neuronal damage induced by aging by exposure to neurotoxicants may be a reflection of highly variable regional energy requirements. This review identifies region-specific vulnerabilities in astrocyte mitochondria in examples of exposure to neurotoxicants and in age-related neurodegeneration.
Collapse
Affiliation(s)
- Laura L Kubik
- Toxicology Program, Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109
| | - Martin A Philbert
- Toxicology Program, Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
36
|
Jia C, Hegg CC. Effect of IP3R3 and NPY on age-related declines in olfactory stem cell proliferation. Neurobiol Aging 2014; 36:1045-56. [PMID: 25482245 DOI: 10.1016/j.neurobiolaging.2014.11.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 10/16/2014] [Accepted: 11/10/2014] [Indexed: 12/18/2022]
Abstract
Losing the sense of smell because of aging compromises health and quality of life. In the mouse olfactory epithelium, aging reduces the capacity for tissue homeostasis and regeneration. The microvillous cell subtype that expresses both inositol trisphosphate receptor type 3 (IP3R3) and the neuroproliferative factor neuropeptide Y (NPY) is critical for regulation of homeostasis, yet its role in aging is undefined. We hypothesized that an age-related decline in IP3R3 expression and NPY signaling underlie age-related homeostatic changes and olfactory dysfunction. We found a decrease in IP3R3(+) and NPY(+) microvillous cell numbers and NPY protein and a reduced sensitivity to NPY-mediated proliferation over 24 months. However, in IP3R3-deficient mice, there was no further age-related reduction in cell numbers, proliferation, or olfactory function compared with wild type. The proliferative response was impaired in aged IP3R3-deficient mice when injury was caused by satratoxin G, which induces IP3R3-mediated NPY release, but not by bulbectomy, which does not evoke NPY release. These data identify IP3R3 and NPY signaling as targets for improving recovery following olfactotoxicant exposure.
Collapse
Affiliation(s)
- Cuihong Jia
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Colleen C Hegg
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
37
|
Chew LJ, DeBoy CA, Senatorov VV. Finding degrees of separation: experimental approaches for astroglial and oligodendroglial cell isolation and genetic targeting. J Neurosci Methods 2014; 236:125-47. [PMID: 25169049 PMCID: PMC4171043 DOI: 10.1016/j.jneumeth.2014.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 08/15/2014] [Accepted: 08/18/2014] [Indexed: 12/20/2022]
Abstract
The study of CNS glial cell function requires experimental methods to detect, purify, and manipulate each cell population with fidelity and specificity. With the identification and cloning of cell- and stage-specific markers, glial cell analysis techniques have grown beyond physical methods of tissue dissociation and cell culture, and become highly specific with immunoselection of cell cultures in vitro and genetic targeting in vivo. The unique plasticity of glial cells offers the potential for cell replacement therapies in neurological disease that utilize neural cells derived from transplanted neural stem and progenitor cells. In this mini-review, we outline general physical and genetic approaches for macroglial cell generation. We summarize cell culture methods to obtain astrocytes and oligodendrocytes and their precursors, from developing and adult tissue, as well as approaches to obtain human neural progenitor cells through the establishment of stem cells. We discuss popular targeting rodent strains designed for cell-specific detection, selection and manipulation of neuroglial cell progenitors and their committed progeny. Based on shared markers between astrocytes and stem cells, we discuss genetically modified mouse strains with overlapping expression, and highlight SOX-expressing strains available for targeting of stem and progenitor cell populations. We also include recently established mouse strains for detection, and tag-assisted RNA and miRNA analysis. This discussion aims to provide a brief overview of the rapidly expanding collection of experimental approaches and genetic resources for the isolation and targeting of macroglial cells, their sources, progeny and gene products to facilitate our understanding of their properties and potential application in pathology.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC, United States.
| | - Cynthia A DeBoy
- Biology Department, Trinity Washington University, Washington, DC, United States
| | - Vladimir V Senatorov
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, United States
| |
Collapse
|
38
|
Are there roles for brain cell senescence in aging and neurodegenerative disorders? Biogerontology 2014; 15:643-60. [PMID: 25305051 DOI: 10.1007/s10522-014-9532-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/13/2014] [Indexed: 12/30/2022]
Abstract
The term cellular senescence was introduced more than five decades ago to describe the state of growth arrest observed in aging cells. Since this initial discovery, the phenotypes associated with cellular senescence have expanded beyond growth arrest to include alterations in cellular metabolism, secreted cytokines, epigenetic regulation and protein expression. Recently, senescence has been shown to play an important role in vivo not only in relation to aging, but also during embryonic development. Thus, cellular senescence serves different purposes and comprises a wide range of distinct phenotypes across multiple cell types. Whether all cell types, including post-mitotic neurons, are capable of entering into a senescent state remains unclear. In this review we examine recent data that suggest that cellular senescence plays a role in brain aging and, notably, may not be limited to glia but also neurons. We suggest that there is a high level of similarity between some of the pathological changes that occur in the brain in Alzheimer's and Parkinson's diseases and those phenotypes observed in cellular senescence, leading us to propose that neurons and glia can exhibit hallmarks of senescence previously documented in peripheral tissues.
Collapse
|
39
|
Shen Y, Gao H, Shi X, Wang N, Ai D, Li J, Ouyang L, Yang J, Tian Y, Lu J. Glutamine synthetase plays a role in d-galactose-induced astrocyte aging in vitro and in vivo. Exp Gerontol 2014; 58:166-73. [DOI: 10.1016/j.exger.2014.08.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 07/31/2014] [Accepted: 08/13/2014] [Indexed: 11/26/2022]
|
40
|
Furman JL, Norris CM. Calcineurin and glial signaling: neuroinflammation and beyond. J Neuroinflammation 2014; 11:158. [PMID: 25199950 PMCID: PMC4172899 DOI: 10.1186/s12974-014-0158-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 08/22/2014] [Indexed: 12/11/2022] Open
Abstract
Similar to peripheral immune/inflammatory cells, neuroglial cells appear to rely on calcineurin (CN) signaling pathways to regulate cytokine production and cellular activation. Several studies suggest that harmful immune/inflammatory responses may be the most impactful consequence of aberrant CN activity in glial cells. However, newly identified roles for CN in glutamate uptake, gap junction regulation, Ca2+ dyshomeostasis, and amyloid production suggest that CN's influence in glia may extend well beyond neuroinflammation. The following review will discuss the various actions of CN in glial cells, with particular emphasis on astrocytes, and consider the implications for neurologic dysfunction arising with aging, injury, and/or neurodegenerative disease.
Collapse
|
41
|
Quincozes-Santos A, Bobermin LD, Tramontina AC, Wartchow KM, Tagliari B, Souza DO, Wyse AT, Gonçalves CA. Oxidative stress mediated by NMDA, AMPA/KA channels in acute hippocampal slices: Neuroprotective effect of resveratrol. Toxicol In Vitro 2014; 28:544-51. [DOI: 10.1016/j.tiv.2013.12.021] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/19/2013] [Accepted: 12/28/2013] [Indexed: 12/19/2022]
|
42
|
Alarcón-Aguilar A, Luna-López A, Ventura-Gallegos JL, Lazzarini R, Galván-Arzate S, González-Puertos VY, Morán J, Santamaría A, Königsberg M. Primary cultured astrocytes from old rats are capable to activate the Nrf2 response against MPP+ toxicity after tBHQ pretreatment. Neurobiol Aging 2014; 35:1901-12. [PMID: 24650792 DOI: 10.1016/j.neurobiolaging.2014.01.143] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 12/18/2013] [Accepted: 01/30/2014] [Indexed: 01/07/2023]
Abstract
Astrocytes are key players for brain physiology, protecting neurons by releasing antioxidant enzymes; however, they are also susceptible to damage by neurotoxins. Nuclear factor erythroid-derived 2-like 2 (Nrf2) is a central regulator of the antioxidant response, and therefore, pharmacologic inducers are often used to activate this transcription factor to induce cellular protection. To date, it still remains unknown if cells from aged animals are capable of developing this response. Therefore, the purpose of this work was to determine if cortical astrocytes derived from old rats are able to respond to tertbuthyl-hydroquinene (tBHQ) pretreatment and stimulate the Nrf2-antioxidant response pathway to induce an antioxidant strategy against MPP+ toxicity, one of the most used molecules to model Parkinson's disease. Our results show that, although astrocytes from adult and old rats were more susceptible to MPP+ toxicity than astrocytes from newborn rats, when pretreated with tertbuthyl-hydroquinene, they were able to transactivate Nrf2, increasing antioxidant enzymes and developing cellular protection. These results are discussed in terms of the doses used to create protective responses.
Collapse
Affiliation(s)
- Adriana Alarcón-Aguilar
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana Iztapalapa, ciudad de México, México
| | - Armando Luna-López
- Area de Ciencia Básica, Instituto Nacional de Geriatría, SSA, ciudad de México, Mexico
| | - José L Ventura-Gallegos
- Departamento de Medicina Genómica y Toxicología Ambiental, IIB, UNAM, ciudad de México, México; Departamento de Bioquímica, INCMNZS, ciudad de México, México
| | - Roberto Lazzarini
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana Iztapalapa, ciudad de México, México
| | - Sonia Galván-Arzate
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, SSA, ciudad de México, México
| | - Viridiana Y González-Puertos
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana Iztapalapa, ciudad de México, México
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, ciudad de México, México
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, SSA, México, México
| | - Mina Königsberg
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana Iztapalapa, ciudad de México, México.
| |
Collapse
|
43
|
Ojeda D, López-Costa JJ, Sede M, López EM, Berria MI, Quarleri J. Increased in vitro glial fibrillary acidic protein expression, telomerase activity, and telomere length after productive human immunodeficiency virus-1 infection in murine astrocytes. J Neurosci Res 2014; 92:267-274. [PMID: 24254728 DOI: 10.1002/jnr.23294] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 08/07/2013] [Accepted: 08/08/2013] [Indexed: 12/17/2022]
Abstract
Although HIV-associated neurocognitive disorders (HAND) result from injury and loss of neurons, productive infection routinely takes place in cells of macrophage lineage. In such a complex context, astrocytosis induced by local chemokines/cytokines is one of the hallmarks of HIV neuropathology. Whether this sustained astrocyte activation is able to alter telomere-aging process is unknown. We hypothesized that interaction of HIV with astrocytes may impact astrocyte telomerase activity (TA) and telomere length in a scenario of astrocytic activation measured by expression of glial fibrillary acidic protein (GFAP). To test this hypothesis, cultured murine astrocytes were challenged with pseudotyped HIV/vesicular stomatitis virus (HIV/VSV) to circumvent the absence of viral receptors; and GFAP, telomerase activity, and telomere length were quantified. As an early and transient event after HIV infection, both TA activity and telomere length were significantly augmented (P < 0.001). Later, a strong negative correlation (-0.8616, P < 0.0001) between virus production and telomerase activity was demonstrated. Once HIV production had reached a peak (7 dpi), the TA decreased, showing levels similar to those of noninfected cells. In contrast, the astrocyte became activated, exhibiting significantly increased levels of GFAP expression directly related to the level of HIV/VSV replication (P < 0.0001). Our results suggest that HIV-infected astrocytes exhibit early disturbance in their cellular functions, such as telomerase activity and telomere length, that may attenuate cell proliferation and enhance the astrocyte dysregulation, contributing to HIV neuropathogenesis. Understanding the mechanisms involved in HIV-mediated persistence by altering the telomere-related aging processes could aid in the development of therapeutic modalities for neurological complications of HIV infection.
Collapse
Affiliation(s)
- Diego Ojeda
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
44
|
Bereiter-Hahn J. Mitochondrial dynamics in aging and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 127:93-131. [PMID: 25149215 DOI: 10.1016/b978-0-12-394625-6.00004-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mitochondria are self-replicating organelles but nevertheless strongly depend on supply coded in nuclear genes. They serve many physiological demands in living cells. Supply of the cytoplasm with ATP and engagement in Ca(2+) regulation belong to the main functions of mitochondria. In large eukaryotic cells, in particular in neurons, with their long dendrites and axons, mitochondria have to move to the sites of their action. This trafficking involves several motor molecules and mechanisms to sense the sites of requirements of mitochondria. With aging and as a consequence of some diseases, mitochondrial components may be rendered dysfunctional, and mtDNA mutations arise during the course of replication and by the action of reactive oxygen species. Mutants in motor molecules engaged in trafficking and in the machinery of fusion and fission are causing severe deficiencies on the cellular level; they support neurodegeneration and, thus, cause many diseases. Frequent fusion and fission events mediate the elimination of impaired parts from mitochondria which finally will be degraded by autophagosomes. Extensive fusion provides a basis for functional complementation. Mobility of proteins and small molecules within the mitochondria is necessary to reach the functional goals of fusion and fission, although cristae and a large fraction of proteins of the respiratory complexes proved to be stable for hours after fusion and perform slow exchange of material.
Collapse
Affiliation(s)
- Jürgen Bereiter-Hahn
- Institute for Cell Biology and Neurosciences, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
45
|
Diekman CO, Fall CP, Lechleiter JD, Terman D. Modeling the neuroprotective role of enhanced astrocyte mitochondrial metabolism during stroke. Biophys J 2013; 104:1752-63. [PMID: 23601322 DOI: 10.1016/j.bpj.2013.02.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 01/29/2013] [Accepted: 02/08/2013] [Indexed: 11/24/2022] Open
Abstract
A mathematical model that integrates the dynamics of cell membrane potential, ion homeostasis, cell volume, mitochondrial ATP production, mitochondrial and endoplasmic reticulum Ca(2+) handling, IP3 production, and GTP-binding protein-coupled receptor signaling was developed. Simulations with this model support recent experimental data showing a protective effect of stimulating an astrocytic GTP-binding protein-coupled receptor (P2Y1Rs) following cerebral ischemic stroke. The model was analyzed to better understand the mathematical behavior of the equations and to provide insights into the underlying biological data. This approach yielded explicit formulas determining how changes in IP3-mediated Ca(2+) release, under varying conditions of oxygen and the energy substrate pyruvate, affected mitochondrial ATP production, and was utilized to predict rate-limiting variables in P2Y1R-enhanced astrocyte protection after cerebral ischemic stroke.
Collapse
Affiliation(s)
- Casey O Diekman
- Mathematical Biosciences Institute, The Ohio State University, Columbus, Ohio, USA.
| | | | | | | |
Collapse
|
46
|
Sama DM, Norris CM. Calcium dysregulation and neuroinflammation: discrete and integrated mechanisms for age-related synaptic dysfunction. Ageing Res Rev 2013; 12:982-95. [PMID: 23751484 PMCID: PMC3834216 DOI: 10.1016/j.arr.2013.05.008] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 05/27/2013] [Accepted: 05/30/2013] [Indexed: 12/30/2022]
Abstract
Some of the best biomarkers of age-related cognitive decline are closely linked to synaptic function and plasticity. This review highlights several age-related synaptic alterations as they relate to Ca(2+) dyshomeostasis, through elevation of intracellular Ca(2+), and neuroinflammation, through production of pro-inflammatory cytokines including interleukin-1 beta (IL-1β) and tumor necrosis factor-alpha (TNF-α). Though distinct in many ways, Ca(2+) and neuroinflammatory signaling mechanisms exhibit extensive cross-talk and bidirectional interactions. For instance, cytokine production in glial cells is strongly dependent on the Ca(2+) dependent protein phosphatase calcineurin, which shows elevated activity in animal models of aging and disease. In turn, pro-inflammatory cytokines, such as TNF, can augment the expression/activity of L-type voltage sensitive Ca(2+) channels in neurons, leading to Ca(2+) dysregulation, hyperactive calcineurin activity, and synaptic depression. Thus, in addition to discussing unique contributions of Ca(2+) dyshomeostasis and neuroinflammation, this review emphasizes how these processes interact to hasten age-related synaptic changes.
Collapse
Affiliation(s)
- Diana M Sama
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | | |
Collapse
|
47
|
P2Y1R-initiated, IP3R-dependent stimulation of astrocyte mitochondrial metabolism reduces and partially reverses ischemic neuronal damage in mouse. J Cereb Blood Flow Metab 2013; 33:600-11. [PMID: 23321785 PMCID: PMC3618399 DOI: 10.1038/jcbfm.2012.214] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glia-based neuroprotection strategies are emerging as promising new avenues to treat brain damage. We previously reported that activation of the glial-specific purinergic receptor, P2Y(1)R, reduces both astrocyte swelling and brain infarcts in a photothrombotic mouse model of stroke. These restorative effects were dependent on astrocyte mitochondrial metabolism. Here, we extend these findings and report that P2Y(1)R stimulation with the purinergic ligand 2-methylthioladenosine 5' diphosphate (2MeSADP) reduces and partially reverses neuronal damage induced by photothrombosis. In vivo neuronal morphology was confocally imaged in transgenic mice expressing yellow fluorescent protein under the control of the Thy1 promoter. Astrocyte mitochondrial membrane potentials, monitored with the potential sensitive dye tetra-methyl rhodamine methyl ester, were depolarized after photothrombosis and subsequently repolarized when P2Y(1)Rs were stimulated. Mice deficient in the astrocyte-specific type 2 inositol 1,4,5 trisphosphate (IP(3)) receptor exhibited aggravated ischemic dendritic damage after photothrombosis. Treatment of these mice with 2MeSADP did not invoke an intracellular Ca(2+) response, did not repolarize astrocyte mitochondria, and did not reduce or partially reverse neuronal lesions induced by photothrombotic stroke. These results demonstrate that IP(3)-Ca(2+) signaling in astrocytes is not only critical for P2Y(1)R-enhanced protection, but suggest that IP(3)-Ca(2+) signaling is also a key component of endogenous neuroprotection.
Collapse
|
48
|
Souza DG, Bellaver B, Souza DO, Quincozes-Santos A. Characterization of adult rat astrocyte cultures. PLoS One 2013; 8:e60282. [PMID: 23555943 PMCID: PMC3610681 DOI: 10.1371/journal.pone.0060282] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 02/24/2013] [Indexed: 11/22/2022] Open
Abstract
Astrocytes, a major class of glial cells, regulate neurotransmitter systems, synaptic processing, ion homeostasis, antioxidant defenses and energy metabolism. Astrocyte cultures derived from rodent brains have been extensively used to characterize astrocytes' biochemical, pharmacological and morphological properties. The aims of this study were to develop a protocol for routine preparation and to characterize a primary astrocyte culture from the brains of adult (90 days old) Wistar rats. For this we used enzymatic digestion (trypsin and papain) and mechanical dissociation. Medium exchange occurred from 24 h after obtaining a culture and after, twice a week up to reach the confluence (around the 4th to 5th week). Under basal conditions, adult astrocytes presented a polygonal to fusiform and flat morphology. Furthermore, approximately 95% the cells were positive for the main glial markers, including GFAP, glutamate transporters, glutamine synthetase and S100B. Moreover, the astrocytes were able to take up glucose and glutamate. Adult astrocytes were also able to respond to acute H2O2 exposure, which led to an increase in reactive oxygen species (ROS) levels and a decrease in glutamate uptake. The antioxidant compound resveratrol was able to protect adult astrocytes from oxidative damage. A response of adult astrocytes to an inflammatory stimulus with LPS was also observed. Changes in the actin cytoskeleton were induced in stimulated astrocytes, most likely by a mechanism dependent on MAPK and Rho A signaling pathways. Taken together, these findings indicate that the culture model described in this study exhibits the biochemical and physiological properties of astrocytes and may be useful for elucidating the mechanisms related to the adult brain, exploring changes between neonatal and adult astrocytes, as well as investigating compounds involved in cytotoxicity and cytoprotection.
Collapse
Affiliation(s)
- Débora Guerini Souza
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | |
Collapse
|
49
|
Contreras-Shannon V, Heart DL, Paredes RM, Navaira E, Catano G, Maffi SK, Walss-Bass C. Clozapine-induced mitochondria alterations and inflammation in brain and insulin-responsive cells. PLoS One 2013; 8:e59012. [PMID: 23527073 PMCID: PMC3604003 DOI: 10.1371/journal.pone.0059012] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 02/09/2013] [Indexed: 01/02/2023] Open
Abstract
Background Metabolic syndrome (MetS) is a constellation of factors including abdominal obesity, hyperglycemia, dyslipidemias, and hypertension that increase morbidity and mortality from diabetes and cardiovascular diseases and affects more than a third of the population in the US. Clozapine, an atypical antipsychotic used for the treatment of schizophrenia, has been found to cause drug-induced metabolic syndrome (DIMS) and may be a useful tool for studying cellular and molecular changes associated with MetS and DIMS. Mitochondria dysfunction, oxidative stress and inflammation are mechanisms proposed for the development of clozapine-related DIMS. In this study, the effects of clozapine on mitochondrial function and inflammation in insulin responsive and obesity-associated cultured cell lines were examined. Methodology/Principal Findings Cultured mouse myoblasts (C2C12), adipocytes (3T3-L1), hepatocytes (FL-83B), and monocytes (RAW 264.7) were treated with 0, 25, 50 and 75 µM clozapine for 24 hours. The mitochondrial selective probe TMRM was used to assess membrane potential and morphology. ATP levels from cell lysates were determined by bioluminescence assay. Cytokine levels in cell supernatants were assessed using a multiplex array. Clozapine was found to alter mitochondria morphology, membrane potential, and volume, and reduce ATP levels in all cell lines. Clozapine also significantly induced the production of proinflammatory cytokines IL-6, GM-CSF and IL12-p70, and this response was particularly robust in the monocyte cell line. Conclusions/Significance Clozapine damages mitochondria and promotes inflammation in insulin responsive cells and obesity-associated cell types. These phenomena are closely associated with changes observed in human and animal studies of MetS, obesity, insulin resistance, and diabetes. Therefore, the use of clozapine in DIMS may be an important and relevant tool for investigating cellular and molecular changes associated with the development of these diseases in the general population.
Collapse
Affiliation(s)
- Verόnica Contreras-Shannon
- Department of Biological Sciences, Saint Mary's University, San Antonio, Texas, United States of America
| | - Dylan L. Heart
- Department of Biological Sciences, Saint Mary's University, San Antonio, Texas, United States of America
| | - R. Madelaine Paredes
- Department of Psychiatry, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Erica Navaira
- Department of Psychiatry, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Gabriel Catano
- Department of Medicine, and the Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Shivani Kaushal Maffi
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas, United States of America
- Medical Research Division, Regional Academic Health Center-Edinburg, Edinburg, Texas, United States of America
| | - Consuelo Walss-Bass
- Department of Psychiatry, University of Texas Health Science Center, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
50
|
Alarcón-Aguilar A, González-Puertos VY, Luna-López A, López-Macay A, Morán J, Santamaría A, Königsberg M. Comparing the effects of two neurotoxins in cortical astrocytes obtained from rats of different ages: involvement of oxidative damage. J Appl Toxicol 2012; 34:127-38. [DOI: 10.1002/jat.2841] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 10/19/2012] [Accepted: 10/20/2012] [Indexed: 01/05/2023]
Affiliation(s)
- Adriana Alarcón-Aguilar
- Departamento de Ciencias de la Salud, DCBS; Universidad Autónoma Metropolitana Iztapalapa; México D.F. 09340 Mexico
| | | | | | - Ambar López-Macay
- Departamento de Ciencias de la Salud, DCBS; Universidad Autónoma Metropolitana Iztapalapa; México D.F. 09340 Mexico
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular; Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | | | - Mina Königsberg
- Departamento de Ciencias de la Salud, DCBS; Universidad Autónoma Metropolitana Iztapalapa; México D.F. 09340 Mexico
| |
Collapse
|