1
|
Ormiston K, Melink Z, Andridge R, Lustberg M, Courtney DeVries A, Murphy K, Emmers K, Ziouzenkova O, Belury MA, Orchard TS. Dietary EPA and DHA enrichment of a high fat diet during doxorubicin-based chemotherapy attenuated neuroinflammatory gene expression in the brain of C57bl/6 ovariectomized mice. Brain Behav Immun 2025; 123:370-382. [PMID: 39313165 DOI: 10.1016/j.bbi.2024.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/28/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024] Open
Abstract
Chemotherapy agents in breast cancer are associated with chemotherapy-related cognitive impairments (CRCI). Mechanisms are not fully clear, but alterations of glucose and lipid metabolism, neuroinflammation and neurodegeneration may contribute to CRCI. The aim of this study was to investigate the combined effects of a high fat (HF) diet combined with doxorubicin-based chemotherapy on glucose and lipid metabolism, neuroinflammation, and neurodegeneration in mice. Additionally, we examined the therapeutic potential of dietary eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) to attenuate these effects. Female C57Bl/6 mice (n = 42) were fed HF, HFn-3 (2 % kcals as EPA + DHA) or Low Fat (LF) diets for seven weeks, with and without chemotherapy. In this study, two chemotherapy injections led to weight and body fat loss associated with a decrease in insulin resistance measured by HOMA-IR. HOMA-IR was significantly greater in HF versus LF groups; but HOMA-IR in HFn-3 group did not significantly differ from either HF or LF groups. Chemotherapy resulted in higher brain concentrations of the inflammatory chemokine KC/GRO. Compared to LF diet plus chemotherapy, HF diet plus chemotherapy upregulated multiple genes involved in neuroinflammation and neurodegeneration pathways. HFn-3 diet plus chemotherapy attenuated gene expression by downregulating multiple genes involved in neuroinflammation and blood brain barrier regulation, including Mapkapk2, Aqp4, and s100b, and upregulating Kcnb1 and Atxn3, genes involved in reduction of oxidative stress and anxiety, respectively. Overall, a HF diet combined with chemotherapy is associated with neuroinflammatory and neurodegenerative gene expression changes in this mouse model; dietary enrichment of EPA and DHA attenuated these effects. Further studies are needed to understand how diet impacts behavioral outcomes of CRCI.
Collapse
Affiliation(s)
- Kate Ormiston
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States
| | - Zihan Melink
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States
| | - Rebecca Andridge
- Division of Biostatistics, College of Public Health, The Ohio State University, United States
| | | | | | - Kelly Murphy
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States
| | - Katie Emmers
- Department of Veterinary Medicine, The Ohio State University, United States
| | - Ouliana Ziouzenkova
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States
| | - Martha A Belury
- Department of Food Science and Technology, The Ohio State University, United States
| | - Tonya S Orchard
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States.
| |
Collapse
|
2
|
Pio-Lopez L, Levin M. Aging as a loss of morphostatic information: A developmental bioelectricity perspective. Ageing Res Rev 2024; 97:102310. [PMID: 38636560 DOI: 10.1016/j.arr.2024.102310] [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: 11/05/2023] [Revised: 02/21/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
Maintaining order at the tissue level is crucial throughout the lifespan, as failure can lead to cancer and an accumulation of molecular and cellular disorders. Perhaps, the most consistent and pervasive result of these failures is aging, which is characterized by the progressive loss of function and decline in the ability to maintain anatomical homeostasis and reproduce. This leads to organ malfunction, diseases, and ultimately death. The traditional understanding of aging is that it is caused by the accumulation of molecular and cellular damage. In this article, we propose a complementary view of aging from the perspective of endogenous bioelectricity which has not yet been integrated into aging research. We propose a view of aging as a morphostasis defect, a loss of biophysical prepattern information, encoding anatomical setpoints used for dynamic tissue and organ homeostasis. We hypothesize that this is specifically driven by abrogation of the endogenous bioelectric signaling that normally harnesses individual cell behaviors toward the creation and upkeep of complex multicellular structures in vivo. Herein, we first describe bioelectricity as the physiological software of life, and then identify and discuss the links between bioelectricity and life extension strategies and age-related diseases. We develop a bridge between aging and regeneration via bioelectric signaling that suggests a research program for healthful longevity via morphoceuticals. Finally, we discuss the broader implications of the homologies between development, aging, cancer and regeneration and how morphoceuticals can be developed for aging.
Collapse
Affiliation(s)
- Léo Pio-Lopez
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA.
| |
Collapse
|
3
|
Su X, Xie L, Li J, Tian X, Lin B, Chen M. Exploring molecular signatures related to the mechanism of aging in different brain regions by integrated bioinformatics. Front Mol Neurosci 2023; 16:1133106. [PMID: 37033380 PMCID: PMC10076559 DOI: 10.3389/fnmol.2023.1133106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/22/2023] [Indexed: 04/11/2023] Open
Abstract
The mechanism of brain aging is not fully understood. Few studies have attempted to identify molecular changes using bioinformatics at the subregional level in the aging brain. This study aimed to identify the molecular signatures and key genes involved in aging, depending on the brain region. Differentially expressed genes (DEGs) associated with aging of the cerebral cortex (CX), hippocampus (HC), and cerebellum (CB) were identified based on five datasets from the Gene Expression Omnibus (GEO). The molecular signatures of aging were explored using functional and pathway analyses. Hub genes of each brain region were determined by protein-protein interaction network analysis, and commonly expressed DEGs (co-DEGs) were also found. Gene-microRNAs (miRNAs) and gene-disease interactions were constructed using online databases. The expression levels and regional specificity of the hub genes and co-DEGs were validated using animal experiments. In total, 32, 293, and 141 DEGs were identified in aging CX, HC, and CB, respectively. Enrichment analysis indicated molecular changes related to leukocyte invasion, abnormal neurotransmission, and impaired neurogenesis due to inflammation as the major signatures of the CX, HC, and CB. Itgax is a hub gene of cortical aging. Zfp51 and Zfp62 were identified as hub genes involved in hippocampal aging. Itgax and Cxcl10 were identified as hub genes involved in cerebellar aging. S100a8 was the only co-DEG in all three regions. In addition, a series of molecular changes associated with inflammation was observed in all three brain regions. Several miRNAs interact with hub genes and S100a8. The change in gene levels was further validated in an animal experiment. Only the upregulation of Zfp51 and Zfp62 was restricted to the HC. The molecular signatures of aging exhibit regional differences in the brain and seem to be closely related to neuroinflammation. Itgax, Zfp51, Zfp62, Cxcl10, and S100a8 may be key genes and potential targets for the prevention of brain aging.
Collapse
Affiliation(s)
- Xie Su
- Department of Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lu Xie
- Department of Physiology, Pre-Clinical Science, Guangxi Medical University, Nanning, China
| | - Jing Li
- Department of Physiology, Pre-Clinical Science, Guangxi Medical University, Nanning, China
| | - Xinyue Tian
- Department of Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bing Lin
- Department of Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Menghua Chen
- Department of Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- *Correspondence: Menghua Chen,
| |
Collapse
|
4
|
Preventing Axonal Sodium Overload or Mitochondrial Calcium Uptake Protects Axonal Mitochondria from Oxidative Stress-Induced Alterations. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6125711. [PMID: 35663200 PMCID: PMC9157283 DOI: 10.1155/2022/6125711] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 11/26/2022]
Abstract
In neuroinflammatory and neurodegenerative disorders such as multiple sclerosis, mitochondrial damage caused by oxidative stress is believed to contribute to neuroaxonal damage. Previously, we demonstrated that exposure to hydrogen peroxide (H2O2) alters mitochondrial morphology and motility in myelinated axons and that these changes initiate at the nodes of Ranvier, where numerous sodium channels are located. Therefore, we suggested that mitochondrial damage may lead to ATP deficit, thereby affecting the efficiency of the sodium-potassium ATPase and eventually leading to sodium overload in axons. The increased intra-axonal sodium may revert the axonal sodium-calcium exchangers and thus may lead to a pathological calcium overload in the axoplasm and mitochondria. Here, we used the explanted murine ventral spinal roots to investigate whether modulation of sodium or calcium influx may prevent mitochondrial alterations in myelinated axons during exogenous application of H2O2 inducing oxidative stress. For that, tetrodotoxin, an inhibitor of voltage-gated sodium ion channels, and ruthenium 360, an inhibitor of the mitochondrial calcium uniporter, were applied simultaneously with hydrogen peroxide to axons. Mitochondrial shape and motility were analyzed. We showed that inhibition of axonal sodium influx prevented oxidative stress-induced morphological changes (i.e., increase in circularity and area and decrease in length) and preserved mitochondrial membrane potential, which is crucial for ATP production. Blocking mitochondrial calcium uptake prevented decrease in mitochondrial motility and also preserved membrane potential. Our findings indicate that alterations of both mitochondrial morphology and motility in the contexts of oxidative stress can be counterbalanced by modulating intramitochondrial ion concentrations pharmacologically. Moreover, motile mitochondria show preserved membrane potentials, pointing to a close association between mitochondrial motility and functionality.
Collapse
|
5
|
Hu X, Jiang Z, Teng L, Yang H, Hong D, Zheng D, Zhao Q. Platinum-Induced Peripheral Neuropathy (PIPN): ROS-Related Mechanism, Therapeutic Agents, and Nanosystems. Front Mol Biosci 2021; 8:770808. [PMID: 34901160 PMCID: PMC8652200 DOI: 10.3389/fmolb.2021.770808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
Platinum (Pt) drugs (e.g., oxaliplatin, cisplatin) are applied in the clinic worldwide for the treatment of various cancers. However, platinum-induced peripheral neuropathy (PIPN) caused by the accumulation of Pt in the peripheral nervous system limits the clinical application, whose prevention and treatment are still a huge challenge. To date, Pt-induced reactive oxygen species (ROS) generation has been studied as one of the primary mechanisms of PIPN, whose downregulation would be feasible to relieve PIPN. This review will discuss ROS-related PIPN mechanisms including Pt accumulation in the dorsal root ganglia (DRG), ROS generation, and cellular regulation. Based on them, some antioxidant therapeutic drugs will be summarized in detail to alleviate the Pt-induced ROS overproduction. More importantly, we focus on the cutting-edge nanotechnology in view of ROS-related PIPN mechanisms and will discuss the rational fabrication of tailor-made nanosystems for efficiently preventing and treating PIPN. Last, the future prospects and potential breakthroughs of these anti-ROS agents and nanosystems will be briefly discussed.
Collapse
Affiliation(s)
- Xi Hu
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijie Jiang
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Longyu Teng
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongyu Yang
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongsheng Hong
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongsheng Zheng
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingwei Zhao
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
6
|
Carvajal FJ, Cerpa W. Regulation of Phosphorylated State of NMDA Receptor by STEP 61 Phosphatase after Mild-Traumatic Brain Injury: Role of Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10101575. [PMID: 34679709 PMCID: PMC8533270 DOI: 10.3390/antiox10101575] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/14/2021] [Accepted: 09/28/2021] [Indexed: 01/21/2023] Open
Abstract
Traumatic Brain Injury (TBI) mediates neuronal death through several events involving many molecular pathways, including the glutamate-mediated excitotoxicity for excessive stimulation of N-methyl-D-aspartate receptors (NMDARs), producing activation of death signaling pathways. However, the contribution of NMDARs (distribution and signaling-associated to the distribution) remains incompletely understood. We propose a critical role of STEP61 (Striatal-Enriched protein tyrosine phosphatase) in TBI; this phosphatase regulates the dephosphorylated state of the GluN2B subunit through two pathways: by direct dephosphorylation of tyrosine-1472 and indirectly via dephosphorylation and inactivation of Fyn kinase. We previously demonstrated oxidative stress’s contribution to NMDAR signaling and distribution using SOD2+/− mice such a model. We performed TBI protocol using a controlled frontal impact device using C57BL/6 mice and SOD2+/− animals. After TBI, we found alterations in cognitive performance, NMDAR-dependent synaptic function (decreased synaptic form of NMDARs and decreased synaptic current NMDAR-dependent), and increased STEP61 activity. These changes are reduced partially with the STEP61-inhibitor TC-2153 treatment in mice subjected to TBI protocol. This study contributes with evidence about the role of STEP61 in the neuropathological progression after TBI and also the alteration in their activity, such as an early biomarker of synaptic damage in traumatic lesions.
Collapse
Affiliation(s)
- Francisco J. Carvajal
- Laboratorio de Función y Patología Neuronal, Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6200000, Chile
| | - Waldo Cerpa
- Laboratorio de Función y Patología Neuronal, Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6200000, Chile
- Correspondence: ; Tel.: +56-2-2354-2656; Fax: +56-2-2354-2660
| |
Collapse
|
7
|
Georgiou CD, Margaritis LH. Oxidative Stress and NADPH Oxidase: Connecting Electromagnetic Fields, Cation Channels and Biological Effects. Int J Mol Sci 2021; 22:10041. [PMID: 34576203 PMCID: PMC8470280 DOI: 10.3390/ijms221810041] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/01/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
Electromagnetic fields (EMFs) disrupt the electrochemical balance of biological membranes, thereby causing abnormal cation movement and deterioration of the function of membrane voltage-gated ion channels. These can trigger an increase of oxidative stress (OS) and the impairment of all cellular functions, including DNA damage and subsequent carcinogenesis. In this review we focus on the main mechanisms of OS generation by EMF-sensitized NADPH oxidase (NOX), the involved OS biochemistry, and the associated key biological effects.
Collapse
Affiliation(s)
- Christos D. Georgiou
- Department of Biology, Section of Genetics, Cell & Developmental Biology, University of Patras, 10679 Patras, Greece;
| | - Lukas H. Margaritis
- Section of Cell Biology and Biophysics, Department of Biology, National and Kapodistrian University of Athens, 26504 Athens, Greece
| |
Collapse
|
8
|
Bruckbauer ST, Minkoff BB, Sussman MR, Cox MM. Proteome Damage Inflicted by Ionizing Radiation: Advancing a Theme in the Research of Miroslav Radman. Cells 2021; 10:cells10040954. [PMID: 33924085 PMCID: PMC8074248 DOI: 10.3390/cells10040954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 02/03/2023] Open
Abstract
Oxidative proteome damage has been implicated as a major contributor to cell death and aging. Protein damage and aging has been a particular theme of the recent research of Miroslav Radman. However, the study of how cellular proteins are damaged by oxidative processes is still in its infancy. Here we examine oxidative changes in the proteomes of four bacterial populations—wild type E. coli, two isolates from E. coli populations evolved for high levels of ionizing radiation (IR) resistance, and D. radiodurans—immediately following exposure to 3000 Gy of ionizing radiation. By a substantial margin, the most prominent intracellular oxidation events involve hydroxylation of methionine residues. Significant but much less frequent are carbonylation events on tyrosine and dioxidation events on tryptophan. A few proteins are exquisitely sensitive to targeted oxidation events, notably the active site of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) in E. coli. Extensive experimental evolution of E. coli for IR resistance has decreased overall proteome sensitivity to oxidation but not to the level seen in D. radiodurans. Many observed oxidation events may reflect aspects of protein structure and/or exposure of protein surfaces to water. Proteins such as GAPDH and possibly Ef-Tu may have an evolved sensitivity to oxidation by H2O2.
Collapse
Affiliation(s)
- Steven T. Bruckbauer
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; (S.T.B.); (M.R.S.)
| | - Benjamin B. Minkoff
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Michael R. Sussman
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; (S.T.B.); (M.R.S.)
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Michael M. Cox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; (S.T.B.); (M.R.S.)
- Correspondence:
| |
Collapse
|
9
|
Dopamine-Dependent QR2 Pathway Activation in CA1 Interneurons Enhances Novel Memory Formation. J Neurosci 2020; 40:8698-8714. [PMID: 33046554 DOI: 10.1523/jneurosci.1243-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 01/18/2023] Open
Abstract
The formation of memory for a novel experience is a critical cognitive capacity. The ability to form novel memories is sensitive to age-related pathologies and disease, to which prolonged metabolic stress is a major contributing factor. Presently, we describe a dopamine-dependent redox modulation pathway within the hippocampus of male mice that promotes memory consolidation. Namely, following novel information acquisition, quinone reductase 2 (QR2) is suppressed by miRNA-182 (miR-182) in the CA1 region of the hippocampus via dopamine D1 receptor (D1R) activation, a process largely facilitated by locus coeruleus activity. This pathway activation reduces ROS generated by QR2 enzymatic activity, a process that alters the intrinsic properties of CA1 interneurons 3 h following learning, in a form of oxidative eustress. Interestingly, novel experience decreases QR2 expression predominately in inhibitory interneurons. Additionally, we find that in aged animals this newly described QR2 pathway is chronically under activated, resulting in miR-182 underexpression and QR2 overexpression. This leads to accumulative oxidative stress, which can be seen in CA1 via increased levels of oxidized, inactivated potassium channel Kv2.1, which undergoes disulfide bridge oligomerization. This newly described interneuron-specific molecular pathway lies alongside the known mRNA translation-dependent processes necessary for long-term memory formation, entrained by dopamine in CA1. It is a process crucial for the distinguishing features of novel memory, and points to a promising new target for memory enhancement in aging and age-dependent diseases.SIGNIFICANCE STATEMENT One way in which evolution dictates which sensory information will stabilize as an internal representation, relies on information novelty. Dopamine is a central neuromodulator involved in this process in the mammalian hippocampus. Here, we describe for the first time a dopamine D1 receptor-dependent quinone reductase 2 pathway in interneurons. This is a targeted redox event necessary to delineate a novel experience to a robust long-term internal representation. Activation of this pathway alone can explain the effect novelty has on "flashbulb" memories, and it can become dysfunctional with age and diseases, such as Alzheimer's disease.
Collapse
|
10
|
Li S, Liu H, Bian S, Sha X, Li Y, Wang Y. The accelerated aging model reveals critical mechanisms of late-onset Parkinson's disease. BioData Min 2020; 13:4. [PMID: 32536974 PMCID: PMC7288517 DOI: 10.1186/s13040-020-00215-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/26/2020] [Indexed: 12/21/2022] Open
Abstract
Background Late-onset Parkinson’s disease (LOPD) is a common neurodegenerative disorder and lacks disease-modifying treatments, attracting major attentions as the aggravating trend of aging population. There were numerous evidences supported that accelerated aging was the primary risk factor for LOPD, thus pointed out that the mechanisms of PD should be revealed thoroughly based on aging acceleration. However, how PD was triggered by accelerated aging remained unclear and the systematic prediction model was needed to study the mechanisms of PD. Results In this paper, an improved PD predictor was presented by comparing with the normal aging process, and both aging and PD markers were identified herein using machine learning methods. Based on the aging scores, the aging acceleration network was constructed thereby, where the enrichment analysis shed light on key characteristics of LOPD. As a result, dysregulated energy metabolisms, the cell apoptosis, neuroinflammation and the ion imbalances were identified as crucial factors linking accelerated aging and PD coordinately, along with dysfunctions in the immune system. Conclusions In short, mechanisms between aging and LOPD were integrated by our computational pipeline.
Collapse
Affiliation(s)
- Shiyan Li
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110122 Liaoning Province China
| | - Hongxin Liu
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110122 Liaoning Province China
| | - Shiyu Bian
- China Medical University, The Queen's University of Belfast Joint College, China Medical University, Shenyang, 110122 Liaoning Province China
| | - Xianzheng Sha
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110122 Liaoning Province China
| | - Yixue Li
- Bio-Med Big Data Center, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Science, Shanghai, 200031 China.,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240 China.,Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, 200433 China.,Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203 China
| | - Yin Wang
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110122 Liaoning Province China.,Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001 Liaoning Province China
| |
Collapse
|
11
|
Muscarinic-Dependent miR-182 and QR2 Expression Regulation in the Anterior Insula Enables Novel Taste Learning. eNeuro 2020; 7:ENEURO.0067-20.2020. [PMID: 32217627 PMCID: PMC7266141 DOI: 10.1523/eneuro.0067-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
In a similar manner to other learning paradigms, intact muscarinic acetylcholine receptor (mAChR) neurotransmission or protein synthesis regulation in the anterior insular cortex (aIC) is necessary for appetitive taste learning. Here we describe a parallel local molecular pathway, where GABAA receptor control of mAChR activation causes upregulation of miRNA-182 and quinone reductase 2 (QR2) mRNA destabilization in the rodent aIC. Damage to long-term memory by prevention of this process, with the use of mAChR antagonist scopolamine before novel taste learning, can be rescued by local QR2 inhibition, demonstrating that QR2 acts downstream of local muscarinic activation. Furthermore, we prove for the first time the presence of endogenous QR2 cofactors in the brain, establishing QR2 as a functional reductase there. In turn, we show that QR2 activity causes the generation of reactive oxygen species, leading to modulation in Kv2.1 redox state. QR2 expression reduction therefore is a previously unaccounted mode of mAChR-mediated inflammation reduction, and thus adds QR2 to the cadre of redox modulators in the brain. The concomitant reduction in QR2 activity during memory consolidation suggests a complementary mechanism to the well established molecular processes of this phase, by which the cortex gleans important information from general sensory stimuli. This places QR2 as a promising new target to tackle neurodegenerative inflammation and the associated impediment of novel memory formation in diseases such as Alzheimer’s disease.
Collapse
|
12
|
Presenilin 1 increases association with synaptotagmin 1 during normal aging. Neurobiol Aging 2019; 86:156-161. [PMID: 31864759 PMCID: PMC7325863 DOI: 10.1016/j.neurobiolaging.2019.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 12/05/2022]
Abstract
Presenilin 1 (PS1), the catalytic component of gamma secretase, associates with synaptotagmin 1 (Syt-1). This interaction is decreased in the brains of patients with sporadic Alzheimer’s disease. However, it remains unclear how this interaction changes during normal aging. Because aging is a risk factor for Alzheimer’s disease, we sought to identify changes in PS1 and Syt-1 association during aging in primary neurons in vitro and mouse brain sections ex vivo. We also tested the effect of aging on the calcium dependence of the interaction by treating neurons aged in vitro with KCl. We found that PS1 and Syt-1 increase their association with age, an effect that is more robust in neuronal processes than cell bodies. Treatment with KCl triggered the interaction in both young and old neurons. Baseline calcium levels and calcium in ux in response to KCl treatment were significantly higher in older neurons, which can partially explain the increase in PS1/Syt-1 binding with age. These results suggest a compensatory mechanism during normal aging to offset detrimental age-associated effects.
Collapse
|
13
|
Yu W, Zhang H, Shin MR, Sesti F. Oxidation of KCNB1 potassium channels in the murine brain during aging is associated with cognitive impairment. Biochem Biophys Res Commun 2019; 512:665-669. [PMID: 30922570 DOI: 10.1016/j.bbrc.2019.03.130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 01/30/2023]
Abstract
Voltage-gated potassium (K+) channel sub-family B member 1 (KCNB1, Kv2.1) is known to undergo oxidation-induced oligomerization during aging but whether this process affects brain's physiology was not known. Here, we used 10, 16 and 22 month-old transgenic mice overexpressing a KCNB1 variant that does not oligomerize (Tg-C73A) and as control, mice overexpressing the wild type (Tg-WT) channel and non-transgenic (non-Tg) mice to elucidate the effects of channel's oxidation on cognitive function. Aging mice in which KCNB1 oligomerization is negligible (Tg-C73A), performed significantly better in the Morris Water Maze (MWM) test of working memory compared to non-Tg or Tg-WT mice. KCNB1 and synapsin-1 co-immunoprecipitated and the cognitive impairment in the MWM was associated with moderate loss of synapsin-1 in pre-synaptic structures of the hippocampus, whereas neurodegeneration and neuronal loss were not significantly different in the various genotypes. We conclude that moderate oxidation of the KCNB1 channel during aging can influence neuronal networks by affecting synaptic function.
Collapse
Affiliation(s)
- Wei Yu
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, 683 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, 683 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Mi Ryung Shin
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, 683 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, 683 Hoes Lane West, Piscataway, NJ, 08854, USA.
| |
Collapse
|
14
|
Cleavage of GSDME by caspase-3 determines lobaplatin-induced pyroptosis in colon cancer cells. Cell Death Dis 2019; 10:193. [PMID: 30804337 PMCID: PMC6389936 DOI: 10.1038/s41419-019-1441-4] [Citation(s) in RCA: 333] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 02/10/2019] [Accepted: 02/11/2019] [Indexed: 02/07/2023]
Abstract
Pyroptosis, a form of programmed cell death (PCD), has garnered increasing attention as it relates to innate immunity and diseases. However, the involvement of pyroptosis in the mechanism by which lobaplatin acts against colorectal cancer (CRC) is unclear. Our study revealed that treatment with lobaplatin reduced the viability of HT-29 and HCT116 cells in a dose-dependent manner. Morphologically, HT-29 and HCT116 cells treated with lobaplatin exhibited microscopic features of cell swelling and large bubbles emerging from the plasma membrane, and transmission electron microscopy (TEM) revealed multiple pores in the membrane. GSDME, rather than GSDMD, was cleaved in lobaplatin-induced pyroptosis in HT-29 and HCT116 cells due to caspase-3 activation. Knocking out GSDME switched lobaplatin-induced cell death from pyroptosis to apoptosis but did not affect lobaplatin-mediated inhibition of growth and tumour formation of HT-29 and HCT116 cells in vivo and in vitro. Further investigation indicates that lobaplatin induced reactive oxygen species (ROS) elevation and JNK phosphorylation. NAC, a ROS scavenger, completely reversed the pyroptosis of lobaplatin-treated HT-29 and HCT116 and JNK phosphorylation. Activated JNK recruited Bax to mitochondria, and thereby stimulated cytochrome c release to cytosol, followed by caspase-3/-9 cleavage and pyroptosis induction. Therefore, in colon cancer cells, GSDME mediates lobaplatin-induced pyroptosis downstream of the ROS/JNK/Bax-mitochondrial apoptotic pathway and caspase-3/-9 activation. Our study indicated that GSDME-dependent pyroptosis is an unrecognized mechanism by which lobaplatin eradicates neoplastic cells, which may have important implications for the clinical application of anticancer therapeutics.
Collapse
|
15
|
Greer JB, Schmale MC, Fieber LA. Whole-transcriptome changes in gene expression accompany aging of sensory neurons in Aplysia californica. BMC Genomics 2018; 19:529. [PMID: 29996779 PMCID: PMC6042401 DOI: 10.1186/s12864-018-4909-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/29/2018] [Indexed: 02/07/2023] Open
Abstract
Background Large-scale molecular changes occur during aging and have many downstream consequences on whole-organism function, such as motor function, learning, and memory. The marine mollusk Aplysia californica can be used to study transcriptional changes that occur with age in identified neurons of the brain, because its simplified nervous system allows for more direct correlations between molecular changes, physiological changes, and their phenotypic outcomes. Behavioral deficits in the tail-withdrawal reflex of aged animals have been correlated with reduced excitation in sensory neurons that control the reflex. RNASeq was used to investigate whole-transcriptome changes in tail-withdrawal sensory neurons of sexually mature and aged Aplysia to correlate transcriptional changes with reduced behavioral and physiological responses. Results Paired-end sequencing resulted in 210 million reads used for differential expression analysis. Aging significantly altered expression of 1202 transcripts in sensory neurons underlying the tail-withdrawal reflex, with an approximately equal number of these genes up- and down regulated with age. Despite overall bidirectionality of expression changes, > 80% of ion channel genes that were differentially expressed had decreased expression with age. In particular, several voltage-gated K+ and Ca2+ channels were down regulated. This marked decrease in ion channel expression may play an important role in previously observed declines in aged sensory neuron excitability. We also observed decreased expression of genes and pathways involved in learning and memory. Genes involved in the stress response showed increased expression in aged Aplysia neurons. Conclusions Significantly altered expression of many genes between sexually mature and aged Aplysia suggests large molecular changes that may impact neuronal function. Decreased ion channel mRNA observed could mean fewer receptors present in aged neurons, resulting in reduced excitability of PVC sensory neurons, ultimately leading to reduced tail-withdrawal reflex observed in aged Aplysia. Significant changes in other genes and pathways, such as stress response and learning and memory, have previously been shown to occur with age in many vertebrate organisms. This suggests that some effects of aging are common across many animal phyla. Electronic supplementary material The online version of this article (10.1186/s12864-018-4909-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Justin B Greer
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, 4600 Rickenbacker Cswy, Miami, FL, 33149, USA.
| | - Michael C Schmale
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, 4600 Rickenbacker Cswy, Miami, FL, 33149, USA
| | - Lynne A Fieber
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, 4600 Rickenbacker Cswy, Miami, FL, 33149, USA
| |
Collapse
|
16
|
Sousa MSB, Holanda IMSD, Monteiro HMC, Amâncio-dos-Santos Â. Antioxidant extract counteracts the effects of aging on cortical spreading depression and oxidative stress in the brain cortex. Acta Cir Bras 2018; 33:472-482. [DOI: 10.1590/s0102-865020180060000001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/12/2018] [Indexed: 11/22/2022] Open
|
17
|
Bothwell MY, Gillette MU. Circadian redox rhythms in the regulation of neuronal excitability. Free Radic Biol Med 2018; 119:45-55. [PMID: 29398284 PMCID: PMC5910288 DOI: 10.1016/j.freeradbiomed.2018.01.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/17/2018] [Accepted: 01/22/2018] [Indexed: 02/07/2023]
Abstract
Oxidation-reduction reactions are essential to life as the core mechanisms of energy transfer. A large body of evidence in recent years presents an extensive and complex network of interactions between the circadian and cellular redox systems. Recent advances show that cellular redox state undergoes a ~24-h (circadian) oscillation in most tissues and is conserved across the domains of life. In nucleated cells, the metabolic oscillation is dependent upon the circadian transcription-translation machinery and, vice versa, redox-active proteins and cofactors feed back into the molecular oscillator. In the suprachiasmatic nucleus (SCN), a hypothalamic region of the brain specialized for circadian timekeeping, redox oscillation was found to modulate neuronal membrane excitability. The SCN redox environment is relatively reduced in daytime when neuronal activity is highest and relatively oxidized in nighttime when activity is at its lowest. There is evidence that the redox environment directly modulates SCN K+ channels, tightly coupling metabolic rhythms to neuronal activity. Application of reducing or oxidizing agents produces rapid changes in membrane excitability in a time-of-day-dependent manner. We propose that this reciprocal interaction may not be unique to the SCN. In this review, we consider the evidence for circadian redox oscillation and its interdependencies with established circadian timekeeping mechanisms. Furthermore, we will investigate the effects of redox on ion-channel gating dynamics and membrane excitability. The susceptibility of many different ion channels to modulation by changes in the redox environment suggests that circadian redox rhythms may play a role in the regulation of all excitable cells.
Collapse
Affiliation(s)
- Mia Y Bothwell
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Martha U Gillette
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
18
|
Carvajal FJ, Mira RG, Rovegno M, Minniti AN, Cerpa W. Age-related NMDA signaling alterations in SOD2 deficient mice. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2010-2020. [PMID: 29577983 DOI: 10.1016/j.bbadis.2018.03.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 12/23/2022]
Abstract
Oxidative stress affects the survival and function of neurons. Hence, they have a complex and highly regulated machinery to handle oxidative changes. The dysregulation of this antioxidant machinery is associated with a wide range of neurodegenerative conditions. Therefore, we evaluated signaling alterations, synaptic properties and behavioral performance in 2 and 6-month-old heterozygous manganese superoxide dismutase knockout mice (SOD2+/- mice). We found that their low antioxidant capacity generated direct oxidative damage in proteins, lipids, and DNA. However, only 6-month-old heterozygous knockout mice presented behavioral impairments. On the other hand, synaptic plasticity, synaptic strength and NMDA receptor (NMDAR) dependent postsynaptic potentials were decreased in an age-dependent manner. We also analyzed the phosphorylation state of the NMDAR subunit GluN2B. We found that while the levels of GluN2B phosphorylated on tyrosine 1472 (synaptic form) remain unchanged, we detected increased levels of GluN2B phosphorylated on tyrosine 1336 (extrasynaptic form), establishing alterations in the synaptic/extrasynaptic ratio of GluN2B. Additionally, we found increased levels of two phosphatases associated with dephosphorylation of p-1472: striatal-enriched protein tyrosine phosphatase (STEP) and phosphatase and tensin homolog deleted on chromosome Ten (PTEN). Moreover, we found decreased levels of p-CREB, a master transcription factor activated by synaptic stimulation. In summary, we describe mechanisms by which glutamatergic synapses are altered under oxidative stress conditions. Our results uncovered new putative therapeutic targets for conditions where NMDAR downstream signaling is altered. This work also contributes to our understanding of processes such as synapse formation, learning, and memory in neuropathological conditions.
Collapse
Affiliation(s)
- Francisco J Carvajal
- Laboratorio de Función y Patología Neuronal, Santiago, Chile; Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Santiago, Chile; Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo G Mira
- Laboratorio de Función y Patología Neuronal, Santiago, Chile; Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Santiago, Chile; Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Maximiliano Rovegno
- Departamento de Medicina Intensiva, Facultad de Medicina, Santiago, Chile; Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alicia N Minniti
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Santiago, Chile; Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Waldo Cerpa
- Laboratorio de Función y Patología Neuronal, Santiago, Chile; Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Santiago, Chile; Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
19
|
Lovisolo D, Dionisi M, A. Ruffinatti F, Distasi C. Nanoparticles and potential neurotoxicity: focus on molecular mechanisms. AIMS MOLECULAR SCIENCE 2018. [DOI: 10.3934/molsci.2018.1.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
20
|
Fukushiro-Lopes DF, Hegel AD, Rao V, Wyatt D, Baker A, Breuer EK, Osipo C, Zartman JJ, Burnette M, Kaja S, Kouzoukas D, Burris S, Jones WK, Gentile S. Preclinical study of a Kv11.1 potassium channel activator as antineoplastic approach for breast cancer. Oncotarget 2017; 9:3321-3337. [PMID: 29423049 PMCID: PMC5790466 DOI: 10.18632/oncotarget.22925] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 11/09/2017] [Indexed: 12/04/2022] Open
Abstract
Potassium ion (K+) channels have been recently found to play a critical role in cancer biology. Despite that pharmacologic manipulation of ion channels is recognized as an important therapeutic approach, very little is known about the effects of targeting of K+ channels in cancer. In this study, we demonstrate that use of the Kv11.1 K+ channel activator NS1643 inhibits tumor growth in an in vivo model of breast cancer. Tumors exposed to NS1643 had reduced levels of proliferation markers, high expression levels of senescence markers, increased production of ROS and DNA damage compared to tumors of untreated mice. Importantly, mice treated with NS1643 did not exhibit significant cardiac dysfunction. In conclusion, pharmacological stimulation of Kv11.1 activity produced arrested TNBC-derived tumor growth by generating DNA damage and senescence without significant side effects. We propose that use of Kv11.1 channels activators could be considered as a possible pharmacological strategy against breast tumors.
Collapse
Affiliation(s)
- Daniela F Fukushiro-Lopes
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Alexandra D Hegel
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Vidhya Rao
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA.,Research Service, Edward Hines Jr. VA Hospital, Hines, IL, USA
| | - Debra Wyatt
- Department of Pathology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Andrew Baker
- Department of Pathology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Eun-Kyoung Breuer
- Department of Pathology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Clodia Osipo
- Department of Pathology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Jeremiah J Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Miranda Burnette
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Simon Kaja
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA.,Department of Ophthalmology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA.,Research Service, Edward Hines Jr. VA Hospital, Hines, IL, USA
| | - Dimitrios Kouzoukas
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Sarah Burris
- Cardiovascular Research Institute, Loyola University Chicago, Maywood, IL, USA
| | - W Keith Jones
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Saverio Gentile
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| |
Collapse
|
21
|
Yan Y, Cheng L, Chen X, Wang Q, Duan M, Ma J, Zhao L, Jiang X, Ai J. Estrogen deficiency is associated with hippocampal morphological remodeling of early postmenopausal mice. Oncotarget 2017; 8:21892-21902. [PMID: 28423534 PMCID: PMC5400632 DOI: 10.18632/oncotarget.15702] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 01/24/2017] [Indexed: 01/10/2023] Open
Abstract
Estrogen (E2) deficiency is reported to involve in the impairment of cognition in postmenopausal women. However, the morphological basis is still unclear. In the present study, using transmission electron microscopy (TEM), we observed the ultrastructure of hippocampus in female C57BL/6 mice at the age of 18 months (18 M) which is considered as the early stage of postmenopause (n = 8). Compared with control mice aged 6 M (n = 8), we identified that the morphological changes in the hippocampus of these menopausal mice were mitochondrial damage, lipofuscin deposition and microtubule degradation. Notably, after E2 was subcutaneously injected into mice aged 16 M with a dosage of 3.5 μg/kg every three days for two months in the 18 M + E2 group (n = 8), mitochondrial damage and lipofuscin deposition in the DG region of hippocampus were prevented, but the degraded microtubules in the hippocampus of postmenopausal mice were failed to restore. These data suggest that hippocampal ultrastructure remodeling in mice can be initiated at the early stage of postmenopause, E2 supplementation could only have an effect on mitochondrial damage and lipofuscin increase.
Collapse
Affiliation(s)
- Yan Yan
- Department of Pharmacology, Harbin Medical University, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Heilongjiang Province, Harbin 150081, China
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Heilongjiang Province, Harbin 150081, China
| | - Xin Chen
- Department of Pharmacology, Harbin Medical University, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Heilongjiang Province, Harbin 150081, China
| | - Qin Wang
- Department of Pharmacology, Harbin Medical University, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Heilongjiang Province, Harbin 150081, China
| | - Mingjing Duan
- Department of Pharmacology, Harbin Medical University, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Heilongjiang Province, Harbin 150081, China
| | - Jichao Ma
- Department of Pharmacology, Harbin Medical University, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Heilongjiang Province, Harbin 150081, China
| | - Linjing Zhao
- Department of Pharmacology, Harbin Medical University, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Heilongjiang Province, Harbin 150081, China
| | - Xuemei Jiang
- Department of Pharmacology, Harbin Medical University, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Heilongjiang Province, Harbin 150081, China
| | - Jing Ai
- Department of Pharmacology, Harbin Medical University, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Heilongjiang Province, Harbin 150081, China
| |
Collapse
|
22
|
Sun Y, Yang PP, Song ZY, Feng Y, Hu DM, Hu J, Xu GY, Zhang HH. α-lipoic acid suppresses neuronal excitability and attenuates colonic hypersensitivity to colorectal distention in diabetic rats. J Pain Res 2017; 10:1645-1655. [PMID: 28769585 PMCID: PMC5529097 DOI: 10.2147/jpr.s135017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AIM Patients with long-standing diabetes often demonstrate intestinal dysfunction, characterized as constipation or colonic hypersensitivity. Our previous studies have demonstrated the roles of voltage-gated sodium channels NaV1.7 and NaV1.8 in dorsal root ganglion (DRG) in colonic hypersensitivity of rats with diabetes. This study was designed to determine roles of antioxidant α-lipoic acid (ALA) on sodium channel activities and colonic hypersensitivity of rats with diabetes. METHODS Streptozotocin was used to induce diabetes in adult female rats. Colonic sensitivity was measured by behavioral responses to colorectal distention in rats. The excitability and sodium channel currents of colon projection DRG neurons labeled with DiI were measured by whole-cell patch-clamp recordings. The expressions of NaV1.7 and NaV1.8 of colon DRGs were measured by western blot analysis. RESULTS ALA treatment significantly increased distention threshold in responding to colorectal distension in diabetic rats compared with normal saline treatment. ALA treatment also hyper-polarized the resting membrane potentials, depolarized action potential threshold, increased rheobase, and decreased frequency of action potentials evoked by ramp current stimulation. Furthermore, ALA treatment also reduced neuronal sodium current densities of DRG neurons innervating the colon from rats with diabetes. In addition, ALA treatment significantly downregulated NaV1.7 and NaV1.8 expression in colon DRGs from rats with diabetes. CONCLUSION Our results suggest that ALA plays an analgesic role, which was likely mediated by downregulation of NaV1.7 and NaV1.8 expressions and functions, thus providing experimental evidence for using ALA to treat colonic hypersensitivity in patients with diabetic visceral pain.
Collapse
Affiliation(s)
- Yan Sun
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Pan-Pan Yang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Zhen-Yuan Song
- Department of Endocrinology, The East District of Suzhou Municipal Hospital, Suzhou, People's Republic of China
| | - Yu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Duan-Min Hu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Guang-Yin Xu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Soochow University, Suzhou, People's Republic of China
| | - Hong-Hong Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
23
|
rhEPO Enhances Cellular Anti-oxidant Capacity to Protect Long-Term Cultured Aging Primary Nerve Cells. J Mol Neurosci 2017. [DOI: 10.1007/s12031-017-0937-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
24
|
Tian K, He CC, Xu HN, Wang YX, Wang HG, An D, Heng B, Pang W, Jiang YG, Liu YQ. Zn 2+ reduction induces neuronal death with changes in voltage-gated potassium and sodium channel currents. J Trace Elem Med Biol 2017; 41:66-74. [PMID: 28347465 DOI: 10.1016/j.jtemb.2017.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 12/27/2016] [Accepted: 02/16/2017] [Indexed: 12/12/2022]
Abstract
In the present study, cultured rat primary neurons were exposed to a medium containing N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine (TPEN), a specific cell membrane-permeant Zn2+ chelator, to establish a model of free Zn2+ deficiency in neurons. The effects of TPEN-mediated free Zn2+ ion reduction on neuronal viability and on the performance of voltage-gated sodium channels (VGSCs) and potassium channels (Kvs) were assessed. Free Zn2+ deficiency 1) markedly reduced the neuronal survival rate, 2) reduced the peak amplitude of INa, 3) shifted the INa activation curve towards depolarization, 4) modulated the sensitivity of sodium channel voltage-dependent inactivation to a depolarization voltage, and 5) increased the time course of recovery from sodium channel inactivation. In addition, free Zn2+ deficiency by TPEN notably enhanced the peak amplitude of transient outward K+ currents (IA) and delayed rectifier K+ currents (IK), as well as caused hyperpolarization and depolarization directional shifts in their steady-state activation curves, respectively. Zn2+ supplementation reversed the effects induced by TPEN. Our results indicate that free Zn2+ deficiency causes neuronal damage and alters the dynamic characteristics of VGSC and Kv currents. Thus, neuronal injury caused by free Zn2+ deficiency may correlate with its modulation of the electrophysiological properties of VGSCs and Kvs.
Collapse
Affiliation(s)
- Kun Tian
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Cong-Cong He
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hui-Nan Xu
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yu-Xiang Wang
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hong-Gang Wang
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Di An
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Bin Heng
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Wei Pang
- Department of Nutrition, Tianjin Institute of Health and Environmental Medicine, Tianjin 300050, China
| | - Yu-Gang Jiang
- Department of Nutrition, Tianjin Institute of Health and Environmental Medicine, Tianjin 300050, China.
| | - Yan-Qiang Liu
- College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
25
|
Ambrogini P, Betti M, Galati C, Di Palma M, Lattanzi D, Savelli D, Galli F, Cuppini R, Minelli A. α-Tocopherol and Hippocampal Neural Plasticity in Physiological and Pathological Conditions. Int J Mol Sci 2016; 17:E2107. [PMID: 27983697 PMCID: PMC5187907 DOI: 10.3390/ijms17122107] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/01/2016] [Accepted: 12/09/2016] [Indexed: 12/25/2022] Open
Abstract
Neuroplasticity is an "umbrella term" referring to the complex, multifaceted physiological processes that mediate the ongoing structural and functional modifications occurring, at various time- and size-scales, in the ever-changing immature and adult brain, and that represent the basis for fundamental neurocognitive behavioral functions; in addition, maladaptive neuroplasticity plays a role in the pathophysiology of neuropsychiatric dysfunctions. Experiential cues and several endogenous and exogenous factors can regulate neuroplasticity; among these, vitamin E, and in particular α-tocopherol (α-T), the isoform with highest bioactivity, exerts potent effects on many plasticity-related events in both the physiological and pathological brain. In this review, the role of vitamin E/α-T in regulating diverse aspects of neuroplasticity is analyzed and discussed, focusing on the hippocampus, a brain structure that remains highly plastic throughout the lifespan and is involved in cognitive functions. Vitamin E-mediated influences on hippocampal synaptic plasticity and related cognitive behavior, on post-natal development and adult hippocampal neurogenesis, as well as on cellular and molecular disruptions in kainate-induced temporal seizures are described. Besides underscoring the relevance of its antioxidant properties, non-antioxidant functions of vitamin E/α-T, mainly involving regulation of cell signaling molecules and their target proteins, have been highlighted to help interpret the possible mechanisms underlying the effects on neuroplasticity.
Collapse
Affiliation(s)
- Patrizia Ambrogini
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Michele Betti
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Claudia Galati
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Michael Di Palma
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Davide Lattanzi
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - David Savelli
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy.
| | - Riccardo Cuppini
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Andrea Minelli
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| |
Collapse
|
26
|
Yu W, Parakramaweera R, Teng S, Gowda M, Sharad Y, Thakker-Varia S, Alder J, Sesti F. Oxidation of KCNB1 Potassium Channels Causes Neurotoxicity and Cognitive Impairment in a Mouse Model of Traumatic Brain Injury. J Neurosci 2016; 36:11084-11096. [PMID: 27798188 PMCID: PMC5098843 DOI: 10.1523/jneurosci.2273-16.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 08/25/2016] [Accepted: 09/07/2016] [Indexed: 01/08/2023] Open
Abstract
The delayed rectifier potassium (K+) channel KCNB1 (Kv2.1), which conducts a major somatodendritic current in cortex and hippocampus, is known to undergo oxidation in the brain, but whether this can cause neurodegeneration and cognitive impairment is not known. Here, we used transgenic mice harboring human KCNB1 wild-type (Tg-WT) or a nonoxidable C73A mutant (Tg-C73A) in cortex and hippocampus to determine whether oxidized KCNB1 channels affect brain function. Animals were subjected to moderate traumatic brain injury (TBI), a condition characterized by extensive oxidative stress. Dasatinib, a Food and Drug Administration-approved inhibitor of Src tyrosine kinases, was used to impinge on the proapoptotic signaling pathway activated by oxidized KCNB1 channels. Thus, typical lesions of brain injury, namely, inflammation (astrocytosis), neurodegeneration, and cell death, were markedly reduced in Tg-C73A and dasatinib-treated non-Tg animals. Accordingly, Tg-C73A mice and non-Tg mice treated with dasatinib exhibited improved behavioral outcomes in motor (rotarod) and cognitive (Morris water maze) assays compared to controls. Moreover, the activity of Src kinases, along with oxidative stress, were significantly diminished in Tg-C73A brains. Together, these data demonstrate that oxidation of KCNB1 channels is a contributing mechanism to cellular and behavioral deficits in vertebrates and suggest a new therapeutic approach to TBI. SIGNIFICANCE STATEMENT This study provides the first experimental evidence that oxidation of a K+ channel constitutes a mechanism of neuronal and cognitive impairment in vertebrates. Specifically, the interaction of KCNB1 channels with reactive oxygen species plays a major role in the etiology of mouse model of traumatic brain injury (TBI), a condition associated with extensive oxidative stress. In addition, a Food and Drug Administration-approved drug ameliorates the outcome of TBI in mouse, by directly impinging on the toxic pathway activated in response to oxidation of the KCNB1 channel. These findings elucidate a basic mechanism of neurotoxicity in vertebrates and might lead to a new therapeutic approach to TBI in humans, which, despite significant efforts, is a condition that remains without effective pharmacological treatments.
Collapse
Affiliation(s)
- Wei Yu
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| | - Randika Parakramaweera
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| | - Shavonne Teng
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| | - Manasa Gowda
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| | - Yashsavi Sharad
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| | - Smita Thakker-Varia
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| | - Janet Alder
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| |
Collapse
|
27
|
Frazier HN, Maimaiti S, Anderson KL, Brewer LD, Gant JC, Porter NM, Thibault O. Calcium's role as nuanced modulator of cellular physiology in the brain. Biochem Biophys Res Commun 2016; 483:981-987. [PMID: 27553276 DOI: 10.1016/j.bbrc.2016.08.105] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/04/2016] [Accepted: 08/18/2016] [Indexed: 12/22/2022]
Abstract
Neuroscientists studying normal brain aging, spinal cord injury, Alzheimer's disease (AD) and other neurodegenerative diseases have focused considerable effort on carefully characterizing intracellular perturbations in calcium dynamics or levels. At the cellular level, calcium is known for controlling life and death and orchestrating most events in between. For many years, intracellular calcium has been recognized as an essential ion associated with nearly all cellular functions from cell growth to degeneration. Often the emphasis is on the negative impact of calcium dysregulation and the typical worse-case-scenario leading inevitably to cell death. However, even high amplitude calcium transients, when executed acutely, can alter neuronal communication and synaptic strength in positive ways, without necessarily killing neurons. Here, we focus on the evidence that calcium has a subtle and distinctive role in shaping and controlling synaptic events that underpin neuronal communication and that these subtle changes in aging or AD may contribute to cognitive decline. We emphasize that calcium imaging in dendritic components is ultimately necessary to directly test for the presence of age- or disease-associated alterations during periods of synaptic activation.
Collapse
Affiliation(s)
- Hilaree N Frazier
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Shaniya Maimaiti
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Katie L Anderson
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Lawrence D Brewer
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - John C Gant
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Nada M Porter
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Olivier Thibault
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA.
| |
Collapse
|