1
|
Ma C, Gou C, Sun S, Wang J, Wei X, Xing F, Xing N, Yuan J, Wang Z. Unraveling the molecular complexity: Wtap/Ythdf1 and Lcn2 in novel traumatic brain injury secondary injury mechanisms. Cell Biol Toxicol 2024; 40:65. [PMID: 39110292 PMCID: PMC11306654 DOI: 10.1007/s10565-024-09909-x] [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: 01/24/2024] [Accepted: 05/15/2024] [Indexed: 08/10/2024]
Abstract
The primary aim of this research was to explore the functions of Wtap and Ythdf1 in regulating neuronal Lipocalin-2 (Lcn2) through m6A modification in traumatic brain injury (TBI). By employing transcriptome sequencing and enrichment analysis, we identified the Wtap/Ythdf1-mediated Lcn2 m6A modification pathway as crucial in TBI. In our in vitro experiments using primary cortical neurons, knockout of Wtap and Ythdf1 led to the inhibition of Lcn2 m6A modification, resulting in reduced neuronal death and inflammation. Furthermore, overexpression of Lcn2 in cortical neurons induced the activation of reactive astrocytes and M1-like microglial cells, causing neuronal apoptosis. In vivo experiments confirmed the activation of reactive astrocytes and microglial cells in TBI and importantly demonstrated that Wtap knockdown improved neuroinflammation and functional impairment. These findings underscore the significance of Wtap/Ythdf1-mediated Lcn2 regulation in TBI secondary injury and suggest potential therapeutic implications for combating TBI-induced neuroinflammation and neuronal damage.
Collapse
Affiliation(s)
- Chaobang Ma
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, 450052, Henan, China
| | - Caili Gou
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Shiyu Sun
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, 450052, Henan, China
| | - Junmin Wang
- Department of Human Anatomy Basic Medical College of Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xin Wei
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Fei Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Jingjing Yuan
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Zhongyu Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, 450052, Henan, China.
- Department of Human Anatomy Basic Medical College of Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
2
|
Zheng ZJ, Zhu LZ, Qiu H, Zheng WYX, You PT, Chen SH, Hu CL, Huang JR, Zhou YJ. Neferine inhibits BMECs pyroptosis and maintains blood-brain barrier integrity in ischemic stroke by triggering a cascade reaction of PGC-1α. Sci Rep 2024; 14:14438. [PMID: 38910141 PMCID: PMC11194274 DOI: 10.1038/s41598-024-64815-w] [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: 01/15/2024] [Accepted: 06/13/2024] [Indexed: 06/25/2024] Open
Abstract
Blood-brain barrier disruption is a critical pathological event in the progression of ischemic stroke (IS). Most studies regarding the therapeutic potential of neferine (Nef) on IS have focused on neuroprotective effect. However, whether Nef attenuates BBB disruption during IS is unclear. We here used mice underwent transient middle cerebral artery occlusion (tMCAO) in vivo and bEnd.3 cells exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) injury in vitro to simulate cerebral ischemia. We showed that Nef reduced neurobehavioral dysfunction and protected brain microvascular endothelial cells and BBB integrity. Molecular docking, short interfering (Si) RNA and plasmid transfection results showed us that PGC-1α was the most binding affinity of biological activity protein for Nef. And verification experiments were showed that Nef upregulated PGC-1α expression to reduce mitochondrial oxidative stress and promote TJ proteins expression, further improves the integrity of BBB in mice. Intriguingly, our study showed that neferine is a natural PGC-1α activator and illustrated the mechanism of specific binding site. Furthermore, we have demonstrated Nef reduced mitochondria oxidative damage and ameliorates endothelial inflammation by inhibiting pyroptosis to improve BBB permeability through triggering a cascade reaction of PGC-1α via regulation of PGC-1α/NLRP3/GSDMD signaling pathway to maintain the integrity of BBB in ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Zi-Jian Zheng
- Department of Pharmacy, Gongan Hospital of Traditional Chinese Medicine, Jingzhou, 434300, China
- Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Li-Zhi Zhu
- Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, 518055, China
| | - Han Qiu
- Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, 3002 West Sungang Rd, Shenzhen, 518020, China
| | - Wu-Yin-Xiao Zheng
- Department of Pharmacy, Gongan Hospital of Traditional Chinese Medicine, Jingzhou, 434300, China
- Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Peng-Tao You
- Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Shu-He Chen
- Department of Pharmacy, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
- Hubei Shizhen Laboratory, Wuhan, 430061, China
| | - Chun-Ling Hu
- Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Jun-Rong Huang
- Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, 518055, China
| | - Ya-Jun Zhou
- Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, 3002 West Sungang Rd, Shenzhen, 518020, China.
- Department of Pharmacy, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China.
- Hubei Shizhen Laboratory, Wuhan, 430061, China.
| |
Collapse
|
3
|
Zou T, Sugimoto K, Zhao Y, Li B, Zhou X, Peng C. Zhi-zi-chi decoction mitigates depression by enhancing lncRNA Six3os1 expression and promoting histone H3K4 methylation at the BDNF promoter. J Cell Mol Med 2024; 28:e18365. [PMID: 38818577 PMCID: PMC11140235 DOI: 10.1111/jcmm.18365] [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: 09/13/2023] [Revised: 03/18/2024] [Accepted: 04/16/2024] [Indexed: 06/01/2024] Open
Abstract
Traditional Chinese medicine, particularly Zhi-zi-chi decoction (ZZCD), is gaining recognition as a potential treatment for depression. This study aimed to uncover the molecular mechanisms behind ZZCD's antidepressant effects, focusing on lncRNA Six3os1 and histone H3K4 methylation at the BDNF promoter. Network pharmacology and in vivo experiments were conducted to identify ZZCD targets and evaluate its impact on depression-related behaviours and neuron injury. The role of Six3os1 in recruiting KMT2A to the BDNF promoter and its effects on oxidative stress and neuron injury were investigated. ZZCD reduced depression-like behaviours and neuron injury in mice subjected to chronic stress. It upregulated Six3os1, which facilitated KMT2A recruitment to the BDNF promoter, leading to increased histone H3K4 methylation and enhanced BDNF expression. ZZCD also inhibited CORT-induced neuron injury, inflammatory response and oxidative stress in vitro. ZZCD's antidepressant properties involve Six3os1 upregulation, which exerts neuroprotective effects by inhibiting oxidative stress and neuron injury, thereby alleviating depressive symptoms. Targeting Six3os1 upregulation may offer a potential therapeutic intervention for depression.
Collapse
Affiliation(s)
- Tianyu Zou
- Department of EncephalopathyShenzhen Luohu District Hospital of Traditional Chinese MedicineShenzhenChina
- Department of EncephalopathyShenzhen Hospital of Shanghai University of Traditional Chinese MedicineShenzhenChina
| | - Kazuo Sugimoto
- Department of Neurology, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
- Institute for Brain DisordersBeijing University of Chinese MedicineBeijingChina
| | - Yu Zhao
- Department of Acupuncture, First Affiliated HospitalHeilongjiang University of Chinese MedicineHarbinChina
| | - Baitao Li
- Department of Acupuncture, First Affiliated HospitalHeilongjiang University of Chinese MedicineHarbinChina
| | - Xiaomao Zhou
- Department of EncephalopathyShenzhen Luohu District Hospital of Traditional Chinese MedicineShenzhenChina
- Department of EncephalopathyShenzhen Hospital of Shanghai University of Traditional Chinese MedicineShenzhenChina
| | - Cheng Peng
- Department of EncephalopathyShenzhen Luohu District Hospital of Traditional Chinese MedicineShenzhenChina
- Department of EncephalopathyShenzhen Hospital of Shanghai University of Traditional Chinese MedicineShenzhenChina
| |
Collapse
|
4
|
Lee H, Kang H, Moon C, Youn B. PAK3 downregulation induces cognitive impairment following cranial irradiation. eLife 2023; 12:RP89221. [PMID: 38131292 PMCID: PMC10746143 DOI: 10.7554/elife.89221] [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] [Indexed: 12/23/2023] Open
Abstract
Cranial irradiation is used for prophylactic brain radiotherapy as well as the treatment of primary brain tumors. Despite its high efficiency, it often induces unexpected side effects, including cognitive dysfunction. Herein, we observed that mice exposed to cranial irradiation exhibited cognitive dysfunction, including altered spontaneous behavior, decreased spatial memory, and reduced novel object recognition. Analysis of the actin cytoskeleton revealed that ionizing radiation (IR) disrupted the filamentous/globular actin (F/G-actin) ratio and downregulated the actin turnover signaling pathway p21-activated kinase 3 (PAK3)-LIM kinase 1 (LIMK1)-cofilin. Furthermore, we found that IR could upregulate microRNA-206-3 p (miR-206-3 p) targeting PAK3. As the inhibition of miR-206-3 p through antagonist (antagomiR), IR-induced disruption of PAK3 signaling is restored. In addition, intranasal administration of antagomiR-206-3 p recovered IR-induced cognitive impairment in mice. Our results suggest that cranial irradiation-induced cognitive impairment could be ameliorated by regulating PAK3 through antagomiR-206-3 p, thereby affording a promising strategy for protecting cognitive function during cranial irradiation, and promoting quality of life in patients with radiation therapy.
Collapse
Affiliation(s)
- Haksoo Lee
- Department of Integrated Biological Science, Pusan National UniversityBusanRepublic of Korea
| | - Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National UniversityBusanRepublic of Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National UniversityGwangjuRepublic of Korea
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National UniversityBusanRepublic of Korea
- Department of Biological Sciences, Pusan National UniversityBusanRepublic of Korea
- Nuclear Science Research Institute, Pusan National UniversityBusanRepublic of Korea
| |
Collapse
|
5
|
Roh J, Kim B, Im M, Jang W, Chae Y, Kang J, Youn B, Kim W. MALAT1-regulated gene expression profiling in lung cancer cell lines. BMC Cancer 2023; 23:818. [PMID: 37667226 PMCID: PMC10476395 DOI: 10.1186/s12885-023-11347-7] [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: 06/14/2023] [Accepted: 08/28/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most common type of lung cancer and has a poor prognosis. Identifying biomarkers based on molecular mechanisms is critical for early diagnosis, timely treatment, and improved prognosis of lung cancer. MALAT1 has been reported to have overexpressed and tumor-promoting functions in NSCLC. It has been proposed as a potential biomarker for the diagnosis and prognosis of cancer. Therefore, this study was conducted to profile the changes in gene expression according to the regulation of expression of MALAT1 in NSCLC cell lines and to investigate the correlation through bioinformatic analysis of differentially expressed genes (DEGs). METHODS MALAT1 expression levels were measured using RT-qPCR. The biological functions of MALAT1 in NSCLC were analyzed by cell counting, colony forming, wound-healing, and Transwell invasion assays. In addition, gene expression profiling in response to the knockdown of MALAT1 was analyzed by transcriptome sequencing, and differentially expressed genes regulated by MALAT1 were performed by GO and KEGG pathway enrichment analyses. Bioinformatic databases were used for gene expression analysis and overall survival analysis. RESULTS Comparative analysis versus MALAT1 expression in MRC5 cells (a normal lung cell line) and the three NSCLC cell lines showed that MALAT1 expression was significantly higher in the NSCLC cells. MALAT1 knockdown decreased cell survival, proliferation, migration, and invasion in all three NSCLC cell lines. RNA-seq analysis of DEGs in NSCLC cells showed 198 DEGs were upregulated and 266 DEGs downregulated by MALAT1 knockdown in all three NSCLC cell lines. Survival analysis on these common DEGs performed using the OncoLnc database resulted in the selection of five DEGs, phosphoglycerate mutase 1 (PGAM1), phosphoglycerate mutase 4 (PGAM4), nucleolar protein 6 (NOL6), nucleosome assembly protein 1 like 5 (NAP1L5), and sestrin1 (SESN1). The gene expression levels of these selected DEGs were proved to gene expression analysis using the TNMplot database. CONCLUSION MALAT1 might function as an oncogene that enhances NSCLC cell survival, proliferation, colony formation, and invasion. RNA-seq and bioinformatic analyses resulted in the selection of five DEGs, PGAM1, PGAM4, NOL6, NAP1L5, and SESN1, which were found to be closely related to patient survival and tumorigenesis. We believe that further investigation of these five DEGs will provide valuable information on the oncogenic role of MALAT1 in NSCLC.
Collapse
Affiliation(s)
- Jungwook Roh
- Department of Science Education, Korea National University of Education, Cheongju-si, 28173, Chungbuk, Republic of Korea
| | - Boseong Kim
- Department of Science Education, Korea National University of Education, Cheongju-si, 28173, Chungbuk, Republic of Korea
| | - Mijung Im
- Department of Science Education, Korea National University of Education, Cheongju-si, 28173, Chungbuk, Republic of Korea
| | - Wonyi Jang
- Department of Science Education, Korea National University of Education, Cheongju-si, 28173, Chungbuk, Republic of Korea
| | - Yeonsoo Chae
- Department of Science Education, Korea National University of Education, Cheongju-si, 28173, Chungbuk, Republic of Korea
| | - JiHoon Kang
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - BuHyun Youn
- Department of Biological Sciences, Pusan National University, Busan, 46241, Republic of Korea
| | - Wanyeon Kim
- Department of Science Education, Korea National University of Education, Cheongju-si, 28173, Chungbuk, Republic of Korea.
- Department of Biology Education, Korea National University of Education, Cheongju-si, 28173, Chungbuk, Republic of Korea.
| |
Collapse
|
6
|
Hardy SJ, Finkelstein AJ, Tivarus M, Culakova E, Mohile N, Weber M, Lin E, Zhong J, Usuki K, Schifitto G, Milano M, Janelsins-Benton MC. Cognitive and neuroimaging outcomes in individuals with benign and low-grade brain tumours receiving radiotherapy: a protocol for a prospective cohort study. BMJ Open 2023; 13:e066458. [PMID: 36792323 PMCID: PMC9933762 DOI: 10.1136/bmjopen-2022-066458] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/27/2023] [Indexed: 02/17/2023] Open
Abstract
INTRODUCTION Radiation-induced cognitive decline (RICD) occurs in 50%-90% of adult patients 6 months post-treatment. In patients with low-grade and benign tumours with long expected survival, this is of paramount importance. Despite advances in radiation therapy (RT) treatment delivery, better understanding of structures important for RICD is necessary to improve cognitive outcomes. We hypothesise that RT may affect network topology and microstructural integrity on MRI prior to any gross anatomical or apparent cognitive changes. In this longitudinal cohort study, we aim to determine the effects of RT on brain structural and functional integrity and cognition. METHODS AND ANALYSIS This study will enroll patients with benign and low-grade brain tumours receiving partial brain radiotherapy. Patients will receive either hypofractionated (>2 Gy/fraction) or conventionally fractionated (1.8-2 Gy/fraction) RT. All participants will be followed for 12 months, with MRIs conducted pre-RT and 6-month and 12 month post-RT, along with a battery of neurocognitive tests and questionnaires. The study was initiated in late 2018 and will continue enrolling through 2024 with final follow-ups completing in 2025. The neurocognitive battery assesses visual and verbal memory, attention, executive function, processing speed and emotional cognition. MRI protocols incorporate diffusion tensor imaging and resting state fMRI to assess structural connectivity and functional connectivity, respectively. We will estimate the association between radiation dose, imaging metrics and cognitive outcomes. ETHICS AND DISSEMINATION This study has been approved by the Research Subjects Review Board at the University of Rochester (STUDY00001512: Cognitive changes in patients receiving partial brain radiation). All results will be published in peer-reviewed journals and at scientific conferences. TRIAL REGISTRATION NUMBER ClinicalTrials.gov NCT04390906.
Collapse
Affiliation(s)
- Sara J Hardy
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Alan J Finkelstein
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
- Center for Advanced Brain Imaging and Neurophysiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Madalina Tivarus
- Center for Advanced Brain Imaging and Neurophysiology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Eva Culakova
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Nimish Mohile
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Miriam Weber
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
| | - Edward Lin
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Jianhui Zhong
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
- Center for Advanced Brain Imaging and Neurophysiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Kenneth Usuki
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
| | - Giovanni Schifitto
- Department of Neurology, Department of Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Michael Milano
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
| | - M C Janelsins-Benton
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
7
|
Gupta U, Baig S, Majid A, Bell SM. The neurology of space flight; How does space flight effect the human nervous system? LIFE SCIENCES IN SPACE RESEARCH 2023; 36:105-115. [PMID: 36682819 DOI: 10.1016/j.lssr.2022.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 06/17/2023]
Abstract
RATIONALE AND HYPOTHESIS Advancements in technology, human adaptability, and funding have increased space exploration and in turn commercial spaceflight. Corporations such as Space X and Blue Origin are exploring methods to make space tourism possible. This could lead to an increase in the number of patients presenting with neurological diseases associated with spaceflight. Therefore, a comprehensive understanding of spaceflight stressors is required to manage neurological disease in high-risk individuals. OBJECTIVES This review aims to describe the neurological effects of spaceflight and to assess countermeasures such as pre-flight prophylaxis, training, and possible therapeutics to reduce long-term effects. METHODOLOGY A literature search was performed for experimental studies conducted in astronauts and in animal models that simulated the space environment. Many studies, however, only discussed these with scientific reasoning and did not include any experimental methods. Relevant studies were identified through searching research databases such as PubMed and Google Scholar. No inclusion or exclusion criteria were used. FINDINGS Analysis of these studies provided a holistic understanding of the acute and chronic neurological changes that occur during space flight. Astronauts are exposed to hazards that include microgravity, cosmic radiation, hypercapnia, isolation, confinement and disrupted circadian rhythms. Microgravity, the absence of a gravitational force, is linked to disturbances in the vestibular system, intracranial and intraocular pressures. Furthermore, microgravity affects near field vision as part of the spaceflight-associated neuro-ocular syndrome. Exposure to cosmic radiation can increase the risk of neurodegenerative conditions and malignancies. It is estimated that cosmic radiation has significantly higher ionising capabilities than the ionising radiation used in medicine. Space travel also has potential benefits to the nervous system, including psychological development and effects on learning and memory. Future work needs to focus on how we can compare a current astronaut to a future space tourist. Potentially the physiological and psychological stresses of space flight might lead to neurological complications in future space travellers that do not have the physiological reserve of current astronauts.
Collapse
Affiliation(s)
- Udit Gupta
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield and S10 2HQ, United Kingdom
| | - Sheharyar Baig
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield and S10 2HQ, United Kingdom; Department of Clinical Neurology, Royal Hallamshire Hospital, Glossop Road, Sheffield, United Kingdom
| | - Arshad Majid
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield and S10 2HQ, United Kingdom; Department of Clinical Neurology, Royal Hallamshire Hospital, Glossop Road, Sheffield, United Kingdom
| | - Simon M Bell
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield and S10 2HQ, United Kingdom; Department of Clinical Neurology, Royal Hallamshire Hospital, Glossop Road, Sheffield, United Kingdom.
| |
Collapse
|
8
|
Profiling of gene expression in the brain associated with anxiety-related behaviors in the chronic phase following cranial irradiation. Sci Rep 2022; 12:13162. [PMID: 35915120 PMCID: PMC9343641 DOI: 10.1038/s41598-022-17310-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 07/22/2022] [Indexed: 11/08/2022] Open
Abstract
Although the brain is exposed to cranial irradiation in many clinical contexts, including malignant brain tumor therapy, such exposure can cause delayed neuropsychiatric disorders in the chronic phase. However, how specific molecular mechanisms are associated with irradiation-induced behavioral dysfunction, especially anxiety-like behaviors, is unclear. In the present study, we evaluated anxiety-like behaviors in adult C57BL/6 mice using the open-field (OF) and elevated plus maze (EPM) tests 3 months following single cranial irradiation (10 Gy). Additionally, by using RNA sequencing (RNA-seq), we analyzed gene expression profiles in the cortex and hippocampus of the adult brain to demonstrate the molecular mechanisms of radiation-induced brain dysfunction. In the OF and EPM tests, mice treated with radiation exhibited increased anxiety-like behaviors in the chronic phase. Gene expression analysis by RNA-seq revealed 89 and 106 differentially expressed genes in the cortex and hippocampus, respectively, following cranial irradiation. Subsequently, ClueGO and STRING analyses clustered these genes in pathways related to protein kinase activity, circadian behavior, and cell differentiation. Based on our expression analysis, we suggest that behavioral dysfunction following cranial irradiation is associated with altered expression of Cdkn1a, Ciart, Fos, Hspa5, Hspb1 and Klf10. These novel findings may provide potential genetic targets to investigate for the development of radioprotective agents.
Collapse
|
9
|
Baiap3 regulates depressive behaviors in mice via attenuating dense core vesicle trafficking in subsets of prefrontal cortex neurons. Neurobiol Stress 2022; 16:100423. [PMID: 35028340 PMCID: PMC8715124 DOI: 10.1016/j.ynstr.2021.100423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 11/21/2022] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are effective first line therapies for treating depression, but are plagued by undesirable side effects and are not effective in all patients. Because SSRIs effectively deplete the neuronal releasable serotonin (5-HT) pool, gaining a deeper understanding of intracellular mechanisms regulating 5-HT pools can help us understand the shortcomings of SSRIs and develop more effective therapies. In this study, we found that BAIAP3 (brain-specific angiogenesis inhibitor 1-associated protein 3) is significantly downregulated in two mouse models of depression (the IR- and CUMS-induced depressive mouse models). In BAIAP3 downregulated models (in vitro and in vivo), we discovered that trafficking of dense core vesicle (DCV), organelles that store, transport and release cargo via exocytosis, was reduced. Accordingly, 5-HT exocytosis and levels in the synapse were lowered, causing defective post-synaptic neurotransmission. In a screen of natural products, we identified eucalyptol, the active components of Eucalyptus, as uniquely capable of increasing neuronal Baiap3 expression and elevate synaptic 5-HT levels. Moreover, eucalyptol treatment relieved depressive behavioral symptoms and restored serotonin levels in mice. Mechanistically, eucalyptol restores Baiap3 expression by reducing inhibitory microRNAs (miR-329, miR-362). These findings illuminate how Baiap3 depletion propagates neurotransmission dysfunction and point to eucalyptol as a novel agent for restoring serotonin exocytosis, suggesting potential for developing eucalyptol as a therapy for treating depression.
Collapse
|
10
|
Early Life Irradiation-Induced Hypoplasia and Impairment of Neurogenesis in the Dentate Gyrus and Adult Depression Are Mediated by MicroRNA- 34a-5p/T-Cell Intracytoplasmic Antigen-1 Pathway. Cells 2021; 10:cells10092476. [PMID: 34572124 PMCID: PMC8466295 DOI: 10.3390/cells10092476] [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: 08/24/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 02/05/2023] Open
Abstract
Early life radiation exposure causes abnormal brain development, leading to adult depression. However, few studies have been conducted to explore pre- or post-natal irradiation-induced depression-related neuropathological changes. Relevant molecular mechanisms are also poorly understood. We induced adult depression by irradiation of mice at postnatal day 3 (P3) to reveal hippocampal neuropathological changes and investigate their molecular mechanism, focusing on MicroRNA (miR) and its target mRNA and protein. P3 mice were irradiated by γ-rays with 5Gy, and euthanized at 1, 7 and 120 days after irradiation. A behavioral test was conducted before the animals were euthanized at 120 days after irradiation. The animal brains were used for different studies including immunohistochemistry, CAP-miRSeq, Real-Time Quantitative Reverse Transcription PCR (qRT-PCR) and western blotting. The interaction of miR-34a-5p and its target T-cell intracytoplasmic antigen-1 (Tia1) was confirmed by luciferase reporter assay. Overexpression of Tia1 in a neural stem cell (NSC) model was used to further validate findings from the mouse model. Irradiation with 5 Gy at P3 induced depression in adult mice. Animal hippocampal pathological changes included hypoplasia of the infrapyramidal blade of the stratum granulosum, aberrant and impaired cell division, and neurogenesis in the dentate gyrus. At the molecular level, upregulation of miR-34a-5p and downregulation of Tia1 mRNA were observed in both animal and neural stem cell models. The luciferase reporter assay and gene transfection studies further confirmed a direct interaction between miR-43a-5p and Tia1. Our results indicate that the early life γ-radiation-activated miR-43a-5p/Tia1 pathway is involved in the pathogenesis of adult depression. This novel finding may provide a new therapeutic target by inhibiting the miR-43a-5p/Tia1 pathway to prevent radiation-induced pathogenesis of depression.
Collapse
|
11
|
Gu YF, Kong LT. Inhibiting p21-activated kinase (PAK7) enhances radiosensitivity in hepatocellular carcinoma. Hum Exp Toxicol 2021; 40:2202-2214. [PMID: 34165002 DOI: 10.1177/09603271211027948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE In light of the upregulation of p21-activated kinase (PAK7) in a variety of cancers, including hepatocellular carcinoma (HCC), we aimed to investigate the effect of PAK7 on the sensitivity of HCC cells to radiotherapy. METHODS PAK7 expression was determined in normal adult liver epithelial THLE-2 and human HCC cell lines. The effect of ionizing radiation (IR) on the HCC cell viability was evaluated by Sulforhodamine B (SRB) assay. HCC cell lines Mahlavu and Huh7 were chosen to assess the effect of PAK7 shRNAs on the viability, clone formation, apoptosis, cycle distribution and γ-H2AX expression after exposure to IR. RESULTS As compared to THLE-2 cells, PAK7 was upregulated in poorly differentiated Mahlavu and SK-Hep-1 cells, but moderately or lowly expressed in well-differentiated Huh7 and HepG2 cells. HCC cells with moderate or low expression of PAK7 presented a decreased viability at 2 Gy IR, which had no significant effect on PAK7high HCC cells. Mahlavu and Huh7 cells transfected with PAK7 shRNAs showed increased inhibitory effect of IR on viability. In addition, PAK7 shRNAs reduced clone formation, facilitated the cell apoptosis, arrested cells at G2/M phase, and increased γ-H2AX expression. Moreover, changes above were more evident in the HCC cells co-treated with IR and PAK7 shRNAs. CONCLUSION PAK7 downregulation could inhibit the viability, promote the apoptosis, arrest cells in G2/M phase, and induce the DNA damage in HCC cells, thereby enhancing the radiosensitivity in HCC.
Collapse
Affiliation(s)
- Y-F Gu
- Department of Radiotherapy Second Ward, YanTaiShan hospital, YanTai, Shandong, People's Republic of China
| | - L-T Kong
- Department of Emergency, YanTaiShan Hospital, YanTai, Shandong, People's Republic of China
| |
Collapse
|
12
|
Zhu JJ, Yu BY, Huang XK, He MZ, Chen BW, Chen TT, Fang HY, Chen SQ, Fu XQ, Li PJ, Lin ZL, Zhu JH. Neferine Protects against Hypoxic-Ischemic Brain Damage in Neonatal Rats by Suppressing NLRP3-Mediated Inflammasome Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6654954. [PMID: 34046147 PMCID: PMC8128543 DOI: 10.1155/2021/6654954] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 04/06/2021] [Accepted: 04/16/2021] [Indexed: 01/06/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is recognized as the main cause of neonatal death, and efficient treatment strategies remain limited. Given the prevalence of HIE and the associated fatality, further studies on its pathogenesis are warranted. Oxidative stress and neuroinflammatory injury are two important factors leading to brain tissue injury and nerve cell loss in HIE. Neferine, an alkaloid extracted from lotus seed embryo, exerts considerable effects against several diseases such as cancers and myocardial injury. In this study, we demonstrated the neuroprotective effect of neferine on HIE and hypothesized that it involves the inhibition of neuronal pyroptosis, thereby ameliorating neurological inflammation and oxidative stress. We demonstrated that the mRNA levels of proteins associated with pyroptosis including caspase-1, the caspase adaptor ASC, gasdermin D, interleukin- (IL-) 18, IL-1β, and some inflammatory factors were significantly increased in neonatal HIBD model rats compared to those in the control group. The increase in these factors was significantly suppressed by treatment with neferine. We stimulated PC12 cells with CoCl2 to induce neuronal HIBD in vitro and investigated the relationship between neferine and pyroptosis by altering the expression of the NLRP3 inflammasome. The overexpression of NLRP3 partially reversed the neuroprotective effect of neferine on HIBD, whereas NLRP3 knockdown further inhibited caspase-1 activation and IL-1β and IL18 expression. In addition, simultaneous alteration of NLRP3 expression induced changes in intracellular oxidative stress levels after HIBD. These findings indicate that neferine ameliorates neuroinflammation and oxidative stress injury by inhibiting pyroptosis after HIBD. Our study provides valuable information for future studies on neferine with respect to neuroinflammation and pyroptosis.
Collapse
Affiliation(s)
- Jin-jin Zhu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Bin-yuan Yu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xiao-kai Huang
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Min-zhi He
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Bin-wen Chen
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Ting-ting Chen
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Huang-yi Fang
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Shang-qin Chen
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xiao-qin Fu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Pei-jun Li
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhen-lang Lin
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jiang-hu Zhu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
13
|
Chen S, Li X, Wu J, Li J, Xiao M, Yang Y, Liu Z, Cheng Y. Plumula Nelumbinis: A review of traditional uses, phytochemistry, pharmacology, pharmacokinetics and safety. JOURNAL OF ETHNOPHARMACOLOGY 2021; 266:113429. [PMID: 33011369 DOI: 10.1016/j.jep.2020.113429] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/06/2020] [Accepted: 09/26/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Plumula Nelumbinis, the green embryo of the mature seeds of Nelumbo nucifera Gaertn, has a medical history of over 400 years. It is widely used for clearing the heart and heat, calming the mind, and promoting astringent essence and hemostasis in traditional Chinese medicine. Moreover, it usually dual use as food and medicine. This review aimed to evaluate the therapeutic potential of Plumula Nelumbinis by summarizing its botany, traditional uses, phytochemistry, pharmacology, pharmacokinetics and safety. METHODS This review summarized published studies on Plumula Nelumbinis in the Chinese Pharmacopoeia and literature databases including PubMed, Web of Science, Baidu Scholar, Wiley and China Knowledge Resource Integrated Database (CNKI), and limits the different research articles in botany, traditional uses, phytochemistry, pharmacology, pharmacokinetics and safety about Plumula Nelumbinis. RESULTS Plumula Nelumbinis is used to treat hypertension, arrhythmia, severe aplastic anemia, insomnia, encephalopathy and gynecological disease in traditional Chinese medicine and clinical studies. More than 130 chemicals have been isolated and identified from Plumula Nelumbinis, including alkaloids, flavonoids, polysaccharides and volatile oil. In addition, pharmacological effects, such as protective effects against cardiovascular diseases, neurological diseases, lung and kidney injury, anti-inflammatory and anticancer activities, were also evaluated by in vitro and in vivo studies. Moreover, the potential signaling pathways regulated by Plumula Nelumbinis in cardiovascular and neurological diseases and perspectives on Plumula Nelumbinis research were discussed. CONCLUSION Plumula Nelumbinis, a commonly used Chinese medicine, has a variety of traditional and modern therapeutic uses. Some traditional uses, especially the treatment of cardiovascular and neurological diseases, have been verified by pharmacological investigation. However, the pharmacological molecular mechanisms, pharmacokinetics and toxicology of Plumula Nelumbinis are still incomplete. In the future, a series of systematic studies on active compounds identification, pharmacological mechanism clarification, quality and safety evaluation are necessary.
Collapse
Affiliation(s)
- Sixuan Chen
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Xuping Li
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Junxuan Wu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Jingyan Li
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Mingzhu Xiao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Ying Yang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Zhongqiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yuanyuan Cheng
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
14
|
Niemietz C, Bezerra F, Almeida MR, Guo S, Monia BP, Saraiva MJ, Schütz P, Schmidt HHJ, Zibert A. SERPINA1 modulates expression of amyloidogenic transthyretin. Exp Cell Res 2020; 395:112217. [PMID: 32768500 DOI: 10.1016/j.yexcr.2020.112217] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 12/14/2022]
Abstract
Hereditary transthyretin amyloidosis (ATTR) is caused by amyloid deposition of misfolded transthyretin (TTR) in various tissues. Recently, reduction of circulating serum TTR, achieved via silencing oligonucleotides, was introduced as therapy of ATTR amyloidosis. We explored the impact of Serpin Family A Member 1 (SERPINA1) on TTR mRNA and protein expression. Oncostatin M (OSM) induced SERPINA1 in hepatoma cells and mice, while concomitantly TTR expression was significantly reduced. SERPINA1 knockdown resulted in specific elevated TTR expression in hepatoma cells; however other genes belonging to the group of acute phase proteins were unaffected. In mice, serum TTR was elevated after mSERPINA1 knockdown throughout antisense treatment. Following SERPINA1 knockdown, TTR deposition in several tissues, including dorsal root ganglia and intestine, was found to be increased, however numbers did not exceed significance levels. The data suggest that SERPINA1 is a co-factor of TTR expression. Our findings provide novel insight in the regulation of TTR and reveal a role of SERPINA1 in the pathogenesis of ATTR amyloidosis.
Collapse
Affiliation(s)
- Christoph Niemietz
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Filipa Bezerra
- i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular,and ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria Rosário Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular,and ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | | | | | - Maria João Saraiva
- i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular,and ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Paula Schütz
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Hartmut H-J Schmidt
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Andree Zibert
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany.
| |
Collapse
|
15
|
Hohsfield LA, Najafi AR, Ghorbanian Y, Soni N, Hingco EE, Kim SJ, Jue AD, Swarup V, Inlay MA, Green KN. Effects of long-term and brain-wide colonization of peripheral bone marrow-derived myeloid cells in the CNS. J Neuroinflammation 2020; 17:279. [PMID: 32951604 PMCID: PMC7504855 DOI: 10.1186/s12974-020-01931-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Microglia, the primary resident myeloid cells of the brain, play critical roles in immune defense by maintaining tissue homeostasis and responding to injury or disease. However, microglial activation and dysfunction has been implicated in a number of central nervous system (CNS) disorders, thus developing tools to manipulate and replace these myeloid cells in the CNS is of therapeutic interest. METHODS Using whole body irradiation, bone marrow transplant, and colony-stimulating factor 1 receptor inhibition, we achieve long-term and brain-wide (~ 80%) engraftment and colonization of peripheral bone marrow-derived myeloid cells (i.e., monocytes) in the brain parenchyma and evaluated the long-term effects of their colonization in the CNS. RESULTS Here, we identify a monocyte signature that includes an upregulation in Ccr1, Ms4a6b, Ms4a6c, Ms4a7, Apobec1, Lyz2, Mrc1, Tmem221, Tlr8, Lilrb4a, Msr1, Nnt, and Wdfy1 and a downregulation of Siglech, Slc2a5, and Ccl21a/b. We demonstrate that irradiation and long-term (~ 6 months) engraftment of the CNS by monocytes induces brain region-dependent alterations in transcription profiles, astrocytes, neuronal structures, including synaptic components, and cognition. Although our results show that microglial replacement with peripherally derived myeloid cells is feasible and that irradiation-induced changes can be reversed by the replacement of microglia with monocytes in the hippocampus, we also observe that brain-wide engraftment of peripheral myeloid cells (relying on irradiation) can result in cognitive and synaptic deficits. CONCLUSIONS These findings provide insight into better understanding the role and complexity of myeloid cells in the brain, including their regulation of other CNS cells and functional outcomes.
Collapse
Affiliation(s)
- Lindsay A Hohsfield
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Allison R Najafi
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Yasamine Ghorbanian
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, 92697, USA
| | - Neelakshi Soni
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Edna E Hingco
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Sung Jin Kim
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Ayer Darling Jue
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Mathew A Inlay
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, 92697, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA.
| |
Collapse
|
16
|
Choi JG, Khan Z, Choi SZ, Kim SY, Oh MS. DA-9801, a standardized Dioscorea extract, improves memory function via the activation of nerve growth factor-mediated signaling. Nutr Neurosci 2020; 25:219-230. [DOI: 10.1080/1028415x.2020.1743916] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Jin Gyu Choi
- Department of Oriental Pharmaceutical Science, College of Pharmacy and Kyung Hee East–West Pharmaceutical Research Institute, Kyung Hee University, Dongdaemun-gu, Republic of Korea
| | - Zahra Khan
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Yeonsu-gu, Republic of Korea
| | | | - Sun Yeou Kim
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Yeonsu-gu, Republic of Korea
| | - Myung Sook Oh
- Department of Oriental Pharmaceutical Science, College of Pharmacy and Kyung Hee East–West Pharmaceutical Research Institute, Kyung Hee University, Dongdaemun-gu, Republic of Korea
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Dongdaemun-gu, Republic of Korea
| |
Collapse
|
17
|
Yang J, Li Q, Han D, Liao C, Wang P, Gao J, Xu Z, Liu Y. Radiation-induced impairment of optic nerve axonal transport in tree shrews and rats monitored by longitudinal manganese-enhanced MRI. Neurotoxicology 2020; 77:145-154. [PMID: 31987859 DOI: 10.1016/j.neuro.2020.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 11/30/2022]
Abstract
PURPOSE Radiation-induced optic neuropathy (RION) is a serious complication that occurs after radiation therapy of tumors in the vicinity of the optic nerve, yet its mechanism and imaging features are poorly understood. In this study, we employed manganese-enhanced MRI (MEMRI) to assess optic nerve axonal transport in tree shrews and rats after irradiation. MATERIALS AND METHODS A comparison of normal visual projections in tree shrews and rats was conducted by intravitreal MnCl2 injection followed by MRI. Adult male tree shrews and rats received a total dose of 20 Gy delivered in two fractions (10 Gy per fraction) within 5 days. Longitudinal MEMRI was conducted 5, 10, 20 and 30 weeks after radiation. At the end of observation, motor proteins involved in axonal transport were detected by western blotting, and the axon cytoskeleton was assessed by immunofluorescence. RESULTS The eyeballs, lens sizes, vitreous volumes, optic nerves and superior colliculi of tree shrews were significantly larger than those of rats on MEMRI (P < 0.05). The Mn2+-enhancement of the optic nerve showed no significant changes at 5 and 10 weeks (P > 0.05) but decreased gradually from 20 to 30 weeks postirradiation (P < 0.05). The enhancement of the superior colliculus gradually decreased from 5 weeks to 30 weeks, and the decrease was most significant at 30 weeks (P < 0.05). The levels of the motor proteins cytoplasmic dynein-1, kinesin-1 and kinesin-2 in the experimental group were significantly decreased (P < 0.05). The immunofluorescence results showed that the α-tubulin, β-tubulin and SMI 31 levels in the experimental groups and control groups were not significantly different (P > 0.05). CONCLUSION Tree shrews show great advantages in visual neuroscience research involving MEMRI. The main cause of the decline in axonal transport in RION is an insufficient level of motor protein rather than damage to the axonal cytoskeletal structure. Longitudinal MEMRI can be used to detect changes in axonal transport function and to observe the relatively intact axon structure from the early to late stages after radiation administration.
Collapse
Affiliation(s)
- Jun Yang
- Department of Radiology. The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital & Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, PR China.
| | - Qinqing Li
- Department of Radiology. The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital & Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, PR China
| | - Dan Han
- Department of Medical Imaging. The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Kunming, 650032, Yunnan, PR China
| | - Chengde Liao
- Department of Radiology. The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital & Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, PR China
| | - Pengfei Wang
- Department of Key Laboratory. The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Road, Kunming, 650101, Yunnan, PR China
| | - Jingyan Gao
- Department of Radiation Oncology. The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital & Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, PR China
| | - Zeyan Xu
- Department of Radiology. The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital & Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, PR China
| | - Yifan Liu
- Department of Radiology. The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital & Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, PR China
| |
Collapse
|
18
|
Xie YC, Yao ZH, Yao XL, Pan JZ, Zhang SF, Zhang Y, Hu JC. Glucagon-Like Peptide-2 Receptor is Involved in Spatial Cognitive Dysfunction in Rats After Chronic Cerebral Hypoperfusion. J Alzheimers Dis 2019; 66:1559-1576. [PMID: 30452417 DOI: 10.3233/jad-180782] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Chronic cerebral hypoperfusion (CCH) affects the aging population and especially patients with neurodegenerative diseases, such as Alzheimer's disease or Parkinson's disease. CCH is closely related to the cognitive dysfunction in these diseases. Glucagon-like peptide-2 receptor (GLP2R) mRNA and protein are highly expressed in the gut and in hippocampal neurons. This receptor is involved in the regulation of food intake and the control of energy balance and glucose homeostasis. The present study employed behavioral techniques, electrophysiology, western blotting, immunohistochemistry, quantitative real time polymerase chain reaction (qRT-PCR), and Golgi staining to investigate whether the expression of GLP2R changes after CCH and whether GLP2R is involved in cognitive impairment caused by CCH. Our findings show that CCH significantly decreased hippocampal GLP2R mRNA and protein levels. GLP2R upregulation could prevent CCH-induced cognitive impairment. It also improved the CCH-induced impairment of long-term potentiation and long-term depression. Additionally, GLP2R modulated after CCH the AKT-mTOR-p70S6K pathway in the hippocampus. Moreover, an upregulation of the GLP2R increased the neurogenesis in the dentate gyrus, neuronal activity, and density of dendritic spines and mushroom spines in hippocampal neurons. Our findings reveal the involvement of GLP2R via a modulation of the AKT-mTOR-p70S6K pathway in the mechanisms underlying CCH-induced impairments of spatial learning and memory. We suggest that the GLP2R and the AKT-mTOR-p70S6K pathway in the hippocampus are promising targets to treat cognition deficits in CCH.
Collapse
Affiliation(s)
- Yan-Chun Xie
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhao-Hui Yao
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiao-Li Yao
- Department of Neurology, Central Hospital of Zhengzhou, Zhengzhou, China
| | - Jian-Zhen Pan
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shao-Feng Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yong Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ji-Chang Hu
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Son B, Lee S, Kim H, Kang H, Kim J, Youn H, Nam SY, Youn B. Low dose radiation attenuates inflammation and promotes wound healing in a mouse burn model. J Dermatol Sci 2019; 96:81-89. [DOI: 10.1016/j.jdermsci.2019.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/28/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023]
|
20
|
Son B, Jeon J, Lee S, Kim H, Kang H, Youn H, Jo S, Youn B. Radiotherapy in combination with hyperthermia suppresses lung cancer progression via increased NR4A3 and KLF11 expression. Int J Radiat Biol 2019; 95:1696-1707. [PMID: 31498019 DOI: 10.1080/09553002.2019.1665213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Purpose: Hyperthermia (HT), a clinical treatment involving delivery of heat to tumors, has been used in combination with traditional chemotherapy and radiotherapy to enhance their effects. However, the molecular mechanism underlying the high efficacy of combination therapy is not clear. This study was conducted to identify the molecular mechanism underlying the sensitization of lung cancer to radiotherapy by HT.Materials and methods: Nuclear receptor subfamily 4, group A, member 3 (NR4A3) and Krüppel-like factor 11 (KLF11) expression in non-small-cell lung cancer cells was confirmed by performing real-time quantitative reverse transcription-polymerase chain reaction. Tumor cell proliferation and apoptosis were assessed via a colony-forming assay and Annexin V/propidium iodide staining.Results and conclusions: Expression profile analysis revealed elevated levels of NR4A3 and KLF11 in A549 lung cancer cells after treatment with HT combined with radiation. We also confirmed that NR4A3 and KLF11 induced apoptosis and inhibited cell proliferation by elevating intracellular reactive oxygen species levels. Knockdown of NR4A3 or KLF11 using siRNA led to decreased effects of radiohyperthermia. Finally, the effect of these two factors on lung cancer progression was evaluated by in vivo xenograft studies. Taken together, the results suggest that NR4A3 and KLF11 are critical for increasing the efficacy of radiotherapy in combination with HT.
Collapse
Affiliation(s)
- Beomseok Son
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea
| | - Jaewan Jeon
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea.,Department of Radiation Oncology, Haeundae Paik Hospital, Inje University School of Medicine, Busan, Republic of Korea
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea
| | - Hyunwoo Kim
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea
| | - Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul, Republic of Korea
| | - Sunmi Jo
- Department of Radiation Oncology, Haeundae Paik Hospital, Inje University School of Medicine, Busan, Republic of Korea
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea.,Department of Biological Sciences, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
21
|
Mariottini C, Munari L, Gunzel E, Seco JM, Tzavaras N, Hansen J, Stern SA, Gao V, Aleyasin H, Sharma A, Azeloglu EU, Hodes GE, Russo SJ, Huff V, Birtwistle MR, Blitzer RD, Alberini CM, Iyengar R. Wilm's tumor 1 promotes memory flexibility. Nat Commun 2019; 10:3756. [PMID: 31434897 PMCID: PMC6704057 DOI: 10.1038/s41467-019-11781-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
Under physiological conditions, strength and persistence of memory must be regulated in order to produce behavioral flexibility. In fact, impairments in memory flexibility are associated with pathologies such as post-traumatic stress disorder or autism; however, the underlying mechanisms that enable memory flexibility are still poorly understood. Here, we identify transcriptional repressor Wilm's Tumor 1 (WT1) as a critical synaptic plasticity regulator that decreases memory strength, promoting memory flexibility. WT1 is activated in the hippocampus following induction of long-term potentiation (LTP) or learning. WT1 knockdown enhances CA1 neuronal excitability, LTP and long-term memory whereas its overexpression weakens memory retention. Moreover, forebrain WT1-deficient mice show deficits in both reversal, sequential learning tasks and contextual fear extinction, exhibiting impaired memory flexibility. We conclude that WT1 limits memory strength or promotes memory weakening, thus enabling memory flexibility, a process that is critical for learning from new experiences.
Collapse
Affiliation(s)
- Chiara Mariottini
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA.
- Systems Biology Center, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA.
| | - Leonardo Munari
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Ellen Gunzel
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
- Systems Biology Center, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Joseph M Seco
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Nikos Tzavaras
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Jens Hansen
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
- Systems Biology Center, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Sarah A Stern
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
- Center for Neural Science, New York University, New York, 10003, NY, USA
| | - Virginia Gao
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
- Center for Neural Science, New York University, New York, 10003, NY, USA
| | - Hossein Aleyasin
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Ali Sharma
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Evren U Azeloglu
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Georgia E Hodes
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Scott J Russo
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Vicki Huff
- Department of Genetics, M.D. Anderson Cancer Center, University of Texas, Houston, 77030, TX, USA
| | - Marc R Birtwistle
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
- Systems Biology Center, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Robert D Blitzer
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Cristina M Alberini
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA.
- Center for Neural Science, New York University, New York, 10003, NY, USA.
| | - Ravi Iyengar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA.
- Systems Biology Center, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA.
| |
Collapse
|
22
|
Do H, Kim D, Kang J, Son B, Seo D, Youn H, Youn B, Kim W. TFAP2C increases cell proliferation by downregulating GADD45B and PMAIP1 in non-small cell lung cancer cells. Biol Res 2019; 52:35. [PMID: 31296259 PMCID: PMC6625030 DOI: 10.1186/s40659-019-0244-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/05/2019] [Indexed: 12/25/2022] Open
Abstract
Background Non-small cell lung cancer (NSCLC) is one of the leading causes of death in the world. NSCLC diagnosed at an early stage can be highly curable with a positive prognosis, but biomarker limitations make it difficult to diagnose lung cancer at an early stage. To identify biomarkers for lung cancer development, we previously focused on the oncogenic roles of transcription factor TFAP2C in lung cancers and revealed the molecular mechanism of several oncogenes in lung tumorigenesis based on TFAP2C-related microarray analysis. Results In this study, we analyzed microarray data to identify tumor suppressor genes and nine genes downregulated by TFAP2C were screened. Among the nine genes, we focused on growth arrest and DNA-damage-inducible beta (GADD45B) and phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1) as representative TFAP2C-regulated tumor suppressor genes. It was observed that overexpressed TFAP2C resulted in inhibition of GADD45B and PMAIP1 expressions at both the mRNA and protein levels in NSCLC cells. In addition, downregulation of GADD45B and PMAIP1 by TFAP2C promoted cell proliferation and cell motility, which are closely associated with NSCLC tumorigenesis. Conclusion This study indicates that GADD45B and PMAIP1 could be promising tumor suppressors for NSCLC and might be useful as prognostic markers for use in NSCLC therapy. Electronic supplementary material The online version of this article (10.1186/s40659-019-0244-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyunhee Do
- Department of Science Education, Korea National University of Education, Cheongju-si, Chungbuk, 28173, Republic of Korea
| | - Dain Kim
- Department of Science Education, Korea National University of Education, Cheongju-si, Chungbuk, 28173, Republic of Korea
| | - JiHoon Kang
- Department of Integrated Biological Science, Pusan National University, Busan, 46241, Republic of Korea
| | - Beomseok Son
- Department of Integrated Biological Science, Pusan National University, Busan, 46241, Republic of Korea
| | - Danbi Seo
- Department of Science Education, Korea National University of Education, Cheongju-si, Chungbuk, 28173, Republic of Korea
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul, 05006, Republic of Korea
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan, 46241, Republic of Korea. .,Department of Biological Sciences, Pusan National University, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea.
| | - Wanyeon Kim
- Department of Science Education, Korea National University of Education, Cheongju-si, Chungbuk, 28173, Republic of Korea. .,Department of Biology Education, Korea National University of Education, 250 Taeseongtabyeon-ro, Gangnae-myeon, Heungdeok-gu, Cheongju-si, Chungbuk, 28173, Republic of Korea.
| |
Collapse
|