1
|
Bai Y, Chang D, Ren H, Ju M, Wang Y, Chen B, Li H, Liu X, Li D, Huo X, Guo X, Tong M, Tan Y, Yao H, Han B. Engagement of N6-methyladenisine methylation of Gng4 mRNA in astrocyte dysfunction regulated by CircHECW2. Acta Pharm Sin B 2024; 14:1644-1660. [PMID: 38572093 PMCID: PMC10985031 DOI: 10.1016/j.apsb.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/08/2023] [Accepted: 01/05/2024] [Indexed: 04/05/2024] Open
Abstract
The N6-methyladenosine (m6A) modification is the most prevalent modification of eukaryotic mRNAs and plays a crucial role in various physiological processes by regulating the stability or function of target mRNAs. Accumulating evidence has suggested that m6A methylation may be involved in the pathological process of major depressive disorder (MDD), a common neuropsychiatric disorder with an unclear aetiology. Here, we found that the levels of the circular RNA HECW2 (circHECW2) were significantly increased in the plasma of both MDD patients and the chronic unpredictable stress (CUS) mouse model. Notably, the downregulation of circHECW2 attenuated astrocyte dysfunction and depression-like behaviors induced by CUS. Furthermore, we demonstrated that the downregulation of circHECW2 increased the expression of the methylase WTAP, leading to an increase in Gng4 expression via m6A modifications. Our findings provide functional insight into the correlation between circHECW2 and m6A methylation, suggesting that circHECW2 may represent a potential target for MDD treatment.
Collapse
Affiliation(s)
- Ying Bai
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Di Chang
- Department of Radiology, Zhongda Hospital, Jiangsu Key Laboratory of Molecular and Functional Imaging, Medical School of Southeast University, Nanjing 210009, China
| | - Hui Ren
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Minzi Ju
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yu Wang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Biling Chen
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Han Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xue Liu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Daxing Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xinchen Huo
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiaofei Guo
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Mengze Tong
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ying Tan
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Honghong Yao
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Bing Han
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
2
|
Alors-Pérez E, Pedraza-Arevalo S, Blázquez-Encinas R, García-Vioque V, Agraz-Doblas A, Yubero-Serrano EM, Sánchez-Frías ME, Serrano-Blanch R, Gálvez-Moreno MÁ, Gracia-Navarro F, Gahete MD, Arjona-Sánchez Á, Luque RM, Ibáñez-Costa A, Castaño JP. Altered CELF4 splicing factor enhances pancreatic neuroendocrine tumors aggressiveness influencing mTOR and everolimus response. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102090. [PMID: 38187140 PMCID: PMC10767201 DOI: 10.1016/j.omtn.2023.102090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 12/01/2023] [Indexed: 01/09/2024]
Abstract
Pancreatic neuroendocrine tumors (PanNETs) comprise a heterogeneous group of tumors with growing incidence. Recent molecular analyses provided a precise picture of their genomic and epigenomic landscape. Splicing dysregulation is increasingly regarded as a novel cancer hallmark influencing key tumor features. We have previously demonstrated that splicing machinery is markedly dysregulated in PanNETs. Here, we aimed to elucidate the molecular and functional implications of CUGBP ELAV-like family member 4 (CELF4), one of the most altered splicing factors in PanNETs. CELF4 expression was determined in 20 PanNETs, comparing tumor and non-tumoral adjacent tissue. An RNA sequencing (RNA-seq) dataset was analyzed to explore CELF4-linked interrelations among clinical features, gene expression, and splicing events. Two PanNET cell lines were employed to assess CELF4 function in vitro and in vivo. PanNETs display markedly upregulated CELF4 expression, which is closely associated with malignancy features, altered expression of key tumor players, and distinct splicing event profiles. Modulation of CELF4 influenced proliferation in vitro and reduced in vivo xenograft tumor growth. Interestingly, functional assays and RNA-seq analysis revealed that CELF4 silencing altered mTOR signaling pathway, enhancing the effect of everolimus. We demonstrate that CELF4 is dysregulated in PanNETs, where it influences tumor development and aggressiveness, likely by modulating the mTOR pathway, suggesting its potential as therapeutic target.
Collapse
Affiliation(s)
- Emilia Alors-Pérez
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Sergio Pedraza-Arevalo
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Ricardo Blázquez-Encinas
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Víctor García-Vioque
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Antonio Agraz-Doblas
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Elena M. Yubero-Serrano
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Unidad de Gestión Clinica Medicina Interna, Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofia University Hospital, Córdoba, Spain
| | - Marina E. Sánchez-Frías
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Pathology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Raquel Serrano-Blanch
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Medical Oncology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - María Ángeles Gálvez-Moreno
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Francisco Gracia-Navarro
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Manuel D. Gahete
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Álvaro Arjona-Sánchez
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- Surgery Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Raúl M. Luque
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Alejandro Ibáñez-Costa
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Justo P. Castaño
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| |
Collapse
|
3
|
Zhu Z, Yang Q, Tian X, Man D, Wang J, Zhang J, Han B. MSTRG3207 promotes apoptosis in zebrafish ZF4 cells via sponging dre-miR-736/bbc3/LOC101885512 axis during cold acclimation. Gene 2024; 894:148010. [PMID: 37981079 DOI: 10.1016/j.gene.2023.148010] [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: 07/13/2023] [Revised: 10/04/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
Long non-coding RNAs (lncRNAs) play essential roles in a variety of biological processes. It has been recently reported that lncRNAs can regulate mRNA expression by binding to microRNAs (miRNAs) as competing endogenous RNAs (ceRNAs). However, the involvement of this regulatory mechanism during cold acclimation in fish remains unclear. In this study, we constructed a ceRNA network mediated by lncRNAs in cold-acclimated zebrafish ZF4 cells through bioinformatic analysis of the mRNA, miRNA, and lncRNA profiles obtained from ZF4 cells cultured at 18 °C for 30 days. A previously uncharacterized lncRNA, MSTRG3207, was selected for further analysis. MSTRG3207 was upregulated and dre-miR-736 was downregulated during cold acclimation. MSTRG3207 was cloned by rapid amplification of cDNA ends (RACE) and functionally characterized. The binding of MSTRG3207 to dre-miR-736 was validated by dual-luciferase reporter assay. Under cold acclimation, MSTRG3207 promoted apoptosis by sponging dre-miR-736 and upregulating bbc3 and LOC101885512, two apoptotic genes targeted by dre-miR-736. Taken together, our findings indicate that MSTRG3207 upregulation promotes apoptosis by sponging dre-miR-736 during cold acclimation in fish.
Collapse
Affiliation(s)
- Zhongqiu Zhu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
| | - Qianting Yang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
| | - Xiaoying Tian
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
| | - Da Man
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
| | - Jian Wang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
| | - Junfang Zhang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, Shanghai 201306, China; Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, China.
| | - Bingshe Han
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, China.
| |
Collapse
|
4
|
Guo X, Wen J, Gao Q, Zhao Y, Zhao Y, Wang C, Xu N, Shao Y, Chang X. Orexin-A/OX1R is involved in regulation of autophagy to promote cortisol secretion in adrenocortical cell. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166844. [PMID: 37572990 DOI: 10.1016/j.bbadis.2023.166844] [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: 02/24/2023] [Revised: 08/03/2023] [Accepted: 08/05/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Hypercortisolism has emerged as a prominent clinical condition worldwide caused by biochemical cortisol excess in patients, and optimization treatment is needed urgently in the clinic. Previously, we observed that orexin-A/orexin type 1 receptor (OX1R) promoted cell proliferation, inhibited apoptosis, and increased cortisol release in adrenocortical cells. However, the functions of orexin-A/OX1R on autophagy and its molecular mechanism are not known. METHODS Transmission electron microscopy and confocal microscope were performed to detect autophagosomes. Western blot were performed to detect autophagy proteins. The cortisol concentration was assessed with an ELISA. FINDINGS Our data demonstrated that orexin-A/OX1R activated the mammalian target of rapamycin/p70 ribosomal protein S6 kinase-1 pathway, thereby inhibiting autophagy in H295R cells and Y-1 cells. Furthermore, the orexin-A/OX1R-mediated suppression of autophagy played a crucial role in cortisol secretion. Mechanistically, the expression of 3β-hydroxysteroid dehydrogenase/isomerase, the rate-limiting enzyme in cortisol synthesis, was increased with autophagy inhibition mediated by orexin-A/OX1R. INTERPRETATION This study provided the evidence that orexin-A/OX1R participated in modulating mTOR/p70S6K1/autophagy signaling pathway to promote cortisol secretion in adrenocortical cell. The findings suggest the mechanistic basis for disorders of cortisol secretion, providing the potential therapeutic targets for hypercortisolism treatment. FUND: This work was supported by National Natural Science Foundation of China (32170603, 31871286), the Doctoral Start-up Foundation of Liaoning Province (20180540008, 2019-BS-298), the Natural Science Foundation of Liaoning Province (2019-ZD-0779), and Shenyang Science and Technology Plan Fund Projects (21-173-9-28).
Collapse
Affiliation(s)
- Xin Guo
- Department of Pediatrics, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Jing Wen
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Qianqian Gao
- Department of the First Obstetric Ward, Wei Fang People's Hospital, Weifang, Shandong 261041, PR China
| | - Yuyan Zhao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110032, PR China
| | - Yue Zhao
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Chunyu Wang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Na Xu
- Natural Sciences Department, LaGuardia Community College (City University of New York), 31-10 Thomson Ave, Long Island City, NY 11101, USA
| | - Yaozhong Shao
- The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shanxi 710061, PR China
| | - Xiaocen Chang
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, PR China.
| |
Collapse
|
5
|
Xie D, Song C, Qin T, Zhai Z, Cai J, Dai J, Sun T, Xu Y. Moschus ameliorates glutamate-induced cellular damage by regulating autophagy and apoptosis pathway. Sci Rep 2023; 13:18586. [PMID: 37903904 PMCID: PMC10616123 DOI: 10.1038/s41598-023-45878-7] [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: 11/16/2022] [Accepted: 10/25/2023] [Indexed: 11/01/2023] Open
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder, causes short-term memory and cognition declines. It is estimated that one in three elderly people die from AD or other dementias. Chinese herbal medicine as a potential drug for treating AD has gained growing interest from many researchers. Moschus, a rare and valuable traditional Chinese animal medicine, was originally documented in Shennong Ben Cao Jing and recognized for its properties of reviving consciousness/resuscitation. Additionally, Moschus has the efficacy of "regulation of menstruation with blood activation, relief of swelling and pain" and is used for treating unconsciousness, stroke, coma, and cerebrovascular diseases. However, it is uncertain whether Moschus has any protective effect on AD patients. We explored whether Moschus could protect glutamate (Glu)-induced PC12 cells from cellular injury and preliminarily explored their related action mechanisms. The chemical compounds of Moschus were analyzed and identified by GC-MS. The Glu-induced differentiated PC12 cell model was thought to be the common AD cellular model. The study aims to preliminarily investigate the intervention effect of Moschus on Glu-induced PC12 cell damage as well as their related action mechanisms. Cell viability, lactate dehydrogenase (LDH), mitochondrial reactive oxygen species, mitochondrial membrane potential (MMP), cell apoptosis, autophagic vacuoles, autolysosomes or autophagosomes, proteins related to apoptosis, and the proteins related to autophagy were examined and analyzed. Seventeen active compounds of the Moschus sample were identified based on GC-MS analysis. In comparison to the control group, Glu stimulation increased cell viability loss, LDH release, mitochondrial damage, loss of MMP, apoptosis rate, and the number of cells containing autophagic vacuoles, and autolysosomes or autophagosomes, while these results were decreased after the pretreatment with Moschus and 3-methyladenine (3-MA). Furthermore, Glu stimulation significantly increased cleaved caspase-3, Beclin1, and LC3II protein expression, and reduced B-cell lymphoma 2/BAX ratio and p62 protein expression, but these results were reversed after pretreatment of Moschus and 3-MA. Moschus has protective activity in Glu-induced PC12 cell injury, and the potential mechanism might involve the regulation of autophagy and apoptosis. Our study may promote research on Moschus in the field of neurodegenerative diseases, and Moschus may be considered as a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Danni Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Caiyou Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhenwei Zhai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jie Cai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jingyi Dai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Ying Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
6
|
Zhang Q, Li G, Kong J, Dai J, Fan Z, Li J. miR‑222-3p reduces neuronal cell apoptosis and alleviates spinal cord injury by inhibiting Bbc3 and Bim. Neurosci Res 2023; 188:39-50. [PMID: 36328305 DOI: 10.1016/j.neures.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
Spinal cord injury (SCI) is a severe traumatic event, but without any established effective treatment because of the irreversible neuronal death. Here, we investigated the role of miR-222-3p in neuronal apoptosis following SCI. Rat SCI models and neuron hypoxia models were accordingly established. The Bbc3, Bim, Bcl-2, Bax, cleaved-caspase 3, cleaved-caspase 9, Cytochrome c, and miR-222-3p expression levels were examined by Western blotting and real-time reverse transcription polymerase chain reaction (RT-qPCR). The possible association between miR-222-3p and Bbc3/Bim was analyzed by dual-luciferase assay. The neuron viability was assessed by Cell Counting Kit-8 assay and Nissl's staining. Live cell staining was performed to detect the mitochondrial membrane potential and neuronal apoptosis. Rat locomotor function was assessed using the Basso-Beattie-Bresnahan scores. Cytochrome c was outflowed from the mitochondria after SCI or hypoxia treatment, and Bbc3, Bim, Bax, cleaved-caspase 9, and cleaved-caspase 3 were significantly upregulated, while Bcl-2 and miR-222-3p were decreased remarkably. Meanwhile, neuronal cell viability was significantly inhibited. Treatment of miR-222-3p significantly suppressed the Cytochrome c efflux and neuronal apoptosis and improved neuronal cell viability and motor function in SCI rats. Moreover, we found that Bbc3 and Bim were the direct targets of miR-222-3p. Overall, our data suggest that miR-222-3p could alleviate the mitochondrial pathway-mediated apoptosis and motor dysfunction in rats after SCI by targeting Bbc3 and Bim.
Collapse
Affiliation(s)
- Qiangqiang Zhang
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Gang Li
- Department of Orthopedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| | - Jundong Kong
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Junyu Dai
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Zhongkai Fan
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China.
| | - Jian Li
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China.
| |
Collapse
|
7
|
Machado da Silva MC, Iglesias LP, Candelario-Jalil E, Khoshbouei H, Moreira FA, de Oliveira ACP. Role of Microglia in Psychostimulant Addiction. Curr Neuropharmacol 2023; 21:235-259. [PMID: 36503452 PMCID: PMC10190137 DOI: 10.2174/1570159x21666221208142151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022] Open
Abstract
The use of psychostimulant drugs can modify brain function by inducing changes in the reward system, mainly due to alterations in dopaminergic and glutamatergic transmissions in the mesocorticolimbic pathway. However, the etiopathogenesis of addiction is a much more complex process. Previous data have suggested that microglia and other immune cells are involved in events associated with neuroplasticity and memory, which are phenomena that also occur in addiction. Nevertheless, how dependent is the development of addiction on the activity of these cells? Although the mechanisms are not known, some pathways may be involved. Recent data have shown psychoactive substances may act directly on immune cells, alter their functions and induce various inflammatory mediators that modulate synaptic activity. These could, in turn, be involved in the pathological alterations that occur in substance use disorder. Here, we extensively review the studies demonstrating how cocaine and amphetamines modulate microglial number, morphology, and function. We also describe the effect of these substances in the production of inflammatory mediators and a possible involvement of some molecular signaling pathways, such as the toll-like receptor 4. Although the literature in this field is scarce, this review compiles the knowledge on the neuroimmune axis that is involved in the pathogenesis of addiction, and suggests some pharmacological targets for the development of pharmacotherapy.
Collapse
Affiliation(s)
- Maria Carolina Machado da Silva
- Department of Pharmacology, Neuropharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil;
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lia Parada Iglesias
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Habibeh Khoshbouei
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fabrício Araujo Moreira
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
8
|
Chen M, Zhang H, Chu YH, Tang Y, Pang XW, Qin C, Tian DS. Microglial autophagy in cerebrovascular diseases. Front Aging Neurosci 2022; 14:1023679. [PMID: 36275005 PMCID: PMC9582432 DOI: 10.3389/fnagi.2022.1023679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Microglia are considered core regulators for monitoring homeostasis in the brain and primary responders to central nervous system (CNS) injuries. Autophagy affects the innate immune functions of microglia. Recently some evidence suggests that microglial autophagy is closely associated with brain function in both ischemic stroke and hemorrhagic stroke. Herein, we will discuss the interaction between autophagy and other biological processes in microglia under physiological and pathological conditions and highlight the interaction between microglial metabolism and autophagy. In the end, we focus on the effect of microglial autophagy in cerebrovascular diseases.
Collapse
|
9
|
Zhao Y, Qin F, Han S, Li S, Zhao Y, Wang H, Tian J, Cen X. MicroRNAs in drug addiction: Current status and future perspectives. Pharmacol Ther 2022; 236:108215. [DOI: 10.1016/j.pharmthera.2022.108215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 12/21/2022]
|
10
|
Deng B, Tang X, Wang Y. Regulation and bioinformatic analysis of circ_0015891/miR-129-1-3p axis in methamphetamine-induced dopaminergic apoptosis. Front Endocrinol (Lausanne) 2022; 13:999211. [PMID: 36204112 PMCID: PMC9530452 DOI: 10.3389/fendo.2022.999211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Methamphetamine (METH) abuse can result in severe neurotoxicity, for which the mechanism is not yet clear. In the present study, we investigated the role of noncoding RNAs in METH-induced dopaminergic neurotoxicity, and analyzed the underlying mechanism using bioinformatic methods. We confirmed by flow cytometry that miR-129-1-3p is involved in promoting dopaminergic apoptosis under METH treatment and its role could be inhibited by a high concentration of circ_0015891. Also, we combined transcriptomic data with bioinformatics to explore the downstream mechanism of miR-129-1-3p regulation of METH-induced apoptosis, highlighted the potentially pivotal figure of response to nutrition. Further bioinformatic analysis of circ_0015891 was conducted as well and showed that circ_0015891 was the sponge of various microRNAs that effect apoptosis by different mechanisms. Collectively, we found a novel circ_0015891/miR-129-1-3p axis that may be a promising therapeutic target for METH-induced dopaminergic neurotoxicity.
Collapse
|
11
|
Liu YP, Cai Y, Lei YD, Yuan XY, Wang Y, Yi S, Li XY, Huang L, Long DX, Zhang ZH. Circular RNA expression profiles in human bronchial epithelial cells treated with beryllium sulfate. Toxicol Res (Camb) 2021; 10:1013-1021. [PMID: 34733486 DOI: 10.1093/toxres/tfab086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/21/2021] [Accepted: 08/05/2021] [Indexed: 11/13/2022] Open
Abstract
Circular RNAs (circRNAs), is a novel type of endogenous non-coding RNAs (ncRNAs) participated in the pathogenesis of many diseases. Beryllium is one of the carcinogenesis elements. However, the mechanism and function of circRNAs in human bronchial epithelial cells (16HBE) induced by beryllium sulfate (BeSO4) was rarely reported. Therefore, the high-throughput RNA sequencing analysis was performed to detect the circRNA profiles between control groups and BeSO4-induced groups. Furthermore, circRNA-miRNA-mRNA network, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and PPI network analysis were used for bioinformatics analysis. CircRNA sequencing analysis revealed that 36 circRNAs were up-regulated and 35 circRNAs were down-regulated in the BeSO4-exposed groups. The selected circRNAs were verified by real-time fluorescent quantitative PCR (qRT-PCR). Hsa_circ_0004214 and hsa_circ_0003586 were validated to be up-regulated, hsa_circ_0047958, hsa_circ_0001944, and hsa_circ_0008982 were down-regulated. The circRNA-miRNA-mRNA network annotated the key signaling pathway including cellular senescence, TNF signaling pathway, NF-kappa B signaling pathway, HIF-1 signaling pathway, and Hippo signaling pathway. The PPI network indicated the most circRNAs might participate in the BeSO4 toxicity by acting as a sponge for the miR-663b through JAK-STAT signaling pathway. In summary, our study suggests that circRNAs may play roles in the mechanism of beryllium toxicity.
Collapse
Affiliation(s)
- Yan-Ping Liu
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Ying Cai
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Yuan-di Lei
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiao-Yan Yuan
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Ye Wang
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Shan Yi
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Xun-Ya Li
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Lian Huang
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Ding-Xin Long
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhao-Hui Zhang
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
12
|
Xu J, Wang X, Wang W, Zhang L, Huang P. Candidate oncogene circularNOP10 mediates gastric cancer progression by regulating miR-204/SIRT1 pathway. J Gastrointest Oncol 2021; 12:1428-1443. [PMID: 34532100 DOI: 10.21037/jgo-21-422] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/17/2021] [Indexed: 12/24/2022] Open
Abstract
Background The role of circular RNA (circRNA) in gastric cancer (GC) is attracting increasing attention. CircNOP10 (hsa_circ-0034351) has been reported to be upregulated in human GC tissue. However, the biological role and mechanism of circNOP10 in GC remain unknown. Methods Circular RNA expression profile of GC was detected based on microarray, and circNOP10 was identified for the subsequent investigation. Clinical samples of GC tissue and patient blood were obtained from the Zhongda Hospital, Southeast University. The different degraded GC cell lines were presented in our laboratory. The function and mechanism of circNOP10 in GC were investigated using Western blot, qRT-PCR, flow cytometry, in situ hybridization and pull down experiment. Results The results indicated that increased circNOP10 in GC tissue was involved in tumor stage and prognosis. In addition, circNOP10 sponged microRNA-24 (miR-204)-mediated biological processes through sirtuin 1 (SIRT1), which further confirmed that the circNOP10/miR-204/SIRT1 pathway promoted proliferation and migration as well as epithelial-mesenchymal transition (EMT) through the NF-κβ pathway in GC cell lines. Conclusions Candidate oncogene circNOP10 mediated GC cell proliferation, arrest cell cycle in G2/M phase, induced cell apoptosis, enhanced tumor metastasis, as well as EMT by activating the miR-204/SIRT1 pathway, suggesting that it may serve as a potential biomarker in GC therapy.
Collapse
Affiliation(s)
- Jiajia Xu
- Department of Clinical Pathology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Xueqing Wang
- Department of Clinical Pathology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Weijie Wang
- Department of Obstet & Gynaecol, Subei Peoples Hospital, Yangzhou, China
| | - Lihua Zhang
- Department of Clinical Pathology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Peilin Huang
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
13
|
Mystery of methamphetamine-induced autophagosome accumulation in hippocampal neurons: loss of syntaxin 17 in defects of dynein-dynactin driving and autophagosome-late endosome/lysosome fusion. Arch Toxicol 2021; 95:3263-3284. [PMID: 34374793 DOI: 10.1007/s00204-021-03131-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 08/04/2021] [Indexed: 01/07/2023]
Abstract
Methamphetamine (METH), a psychoactive-stimulant facilitates massive accumulation of autophagosomes and causes autophagy-associated neuronal death. However, the underlying mechanisms involving METH-induced auto-phagosome accumulation remain poorly understood. In the current study, autophagic flux was tracked by mRFP-GFP-LC3 adenovirus, 900 μM METH treatment was found to significantly disrupt autophagic flux, which was further validated by remarkable increase of co-localized of LC3 and SQSTM1/p62, enhancement of LC3-II and SQSTM1/p62 protein levels, and massive autophagosome puncta aggregation. With the cycloheximide (CHX) treatment, METH treatment was displayed a significant inhibition of SQSTM1/p62 degradation. Therefore, the mRNAs associated with vesicle degradation were screened, and syntaxin 17 (Stx17) and dynein-dynactin mRNA levels significantly decreased, an effect was proved in protein level as well. Intriguingly, METH induced autophagosome accumulation and autophagic flux disturbance was incredibly retarded by overexpression of Stx17, which was validated by the restoration of the fusion autophagosome-late endosome/lysosome fusion. Moreover, Stx17 overexpression obviously impeded the METH-induced decrease of co-localization of the retrograded motor protein dynein/dynactin and autophagosome-late endosome, though the dynein/dynactin proteins were not involved in autophagosome-late endosome/lysosome fusion. Collectively, our findings unravel the mechanism of METH-induced autophagosome accumulation involving autophagosome-late endosome/lysosome fusion deficiency and that autophagy-enhancing mechanisms such as the overexpression of Stx17 may be therapeutic strategies for the treatment of METH-induced neuronal damage.
Collapse
|
14
|
Pathological methamphetamine exposure triggers the accumulation of neuropathic protein amyloid-β by inhibiting UCHL1. Neurotoxicology 2021; 86:19-25. [PMID: 34175320 DOI: 10.1016/j.neuro.2021.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/10/2021] [Accepted: 06/22/2021] [Indexed: 11/21/2022]
Abstract
Methamphetamine (METH), a powerful psychoactive drug, causes damage to the nervous system and leads to degenerative changes similar to Alzheimer's disease (AD), however, the molecular mechanism between the toxicity of METH and AD-related symptoms remains poorly understood. In this study, we investigated the effect of METH exposure on the accumulation of amyloid-β by establishing the animal and cell models. The results showed that METH exposure increased amyloid precursor protein (APP) and β-secretase (BACE1), contributed to the accumulation of amyloid-β, and which was alleviated with the pretreatment of BACE1 inhibitor. In addition, METH exposure decreased ubiquitin carboxy-terminal hydrolases L1 (UCHL1) which was related to the degradation of BACE1, and therefore led to the up-regulation of BACE1. In summary, the study could provide a new insight into the molecular mechanisms of METH toxicity and new evidence for the link between METH abuse and AD.
Collapse
|
15
|
Non-coding RNA: insights into the mechanism of methamphetamine neurotoxicity. Mol Cell Biochem 2021; 476:3319-3328. [PMID: 33895910 DOI: 10.1007/s11010-021-04160-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 04/15/2021] [Indexed: 10/21/2022]
Abstract
Chronic exposure of the methamphetamine has been shown to lead to neurotoxicity in rodents and humans. The manifestations of methamphetamine neurotoxicity include methamphetamine use disorder, methamphetamine abuse, methamphetamine addiction and methamphetamine behavioral sensitization. Repeated use of methamphetamine can cause methamphetamine use disorder. The abuse and addiction of methamphetamine are growing epidemic worldwide. Repeated intermittent exposure to methamphetamine can cause behavioral sensitization. In addition, many studies have shown that changes in the expression of non-coding RNA in the ventral tegmental area and nucleus accumbens will affect the behavioral effects of methamphetamine. Non-coding RNA plays an important role in the behavioral effects of methamphetamine. Therefore, it is important to study the relationship between methamphetamine and non-coding RNA. The purpose of this review is to study the non-coding RNA associated with methamphetamine neurotoxicity to search for the possible therapeutic target of the methamphetamine neurotoxicity.
Collapse
|
16
|
Smooth muscle-specific HuR knockout induces defective autophagy and atherosclerosis. Cell Death Dis 2021; 12:385. [PMID: 33837179 PMCID: PMC8035143 DOI: 10.1038/s41419-021-03671-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 01/19/2023]
Abstract
Human antigen R (HuR) is a widespread RNA-binding protein involved in homeostatic regulation and pathological processes in many diseases. Atherosclerosis is the leading cause of cardiovascular disease and acute cardiovascular events. However, the role of HuR in atherosclerosis remains unknown. In this study, mice with smooth muscle-specific HuR knockout (HuRSMKO) were generated to investigate the role of HuR in atherosclerosis. HuR expression was reduced in atherosclerotic plaques. As compared with controls, HuRSMKO mice showed increased plaque burden in the atherosclerotic model. Mechanically, HuR could bind to the mRNAs of adenosine 5′-monophosphate-activated protein kinase (AMPK) α1 and AMPKα2, thus increasing their stability and translation. HuR deficiency reduced p-AMPK and LC3II levels and increased p62 level, thereby resulting in defective autophagy. Finally, pharmacological AMPK activation induced autophagy and suppressed atherosclerosis in HuRSMKO mice. Our findings suggest that smooth muscle HuR has a protective effect against atherosclerosis by increasing AMPK-mediated autophagy.
Collapse
|
17
|
Hadizadeh-Bazaz M, Vaezi G, Khaksari M, Hojati V. Curcumin attenuates spatial memory impairment by anti-oxidative, anti-apoptosis, and anti-inflammatory mechanism against methamphetamine neurotoxicity in male Wistar rats: Histological and biochemical changes. Neurotoxicology 2021; 84:208-217. [PMID: 33819551 DOI: 10.1016/j.neuro.2021.03.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Methamphetamine is used extensively around the world as a psychostimulant. The complications related to methamphetamine include methamphetamine-induced neurotoxicity, mainly involving intraneuronal processes, such as oxidative stress and excitotoxicity. Curcumin is effective against neuronal injury due to its antioxidant, anti-inflammatory effects. In this study, we examined the protective effects of curcumin against methamphetamine neurotoxicity. METHODS Sixty male Wistar rats were divided into the following groups: control (n = 12), DMSO (n = 12), methamphetamine (n = 12), and methamphetamine + curcumin (100 and 200 mg/kg, respectively, intraperitoneal [IP]; n = 12). Neurotoxicity was induced by 40 mg/kg of methamphetamine administrated through 4 injections (4 × 10 mg/kg, q2h, IP). Curcumin (100 and 200 mg/kg) was administered at 7 days after the last methamphetamine injection. By using a Morris water maze task, the hippocampus-dependent memory and spatial learning were evaluated 1 day after the last curcumin injection. Then, the animal brains were isolated for biochemical measurements, as well as glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor protein-1(Iba-1) and caspase-3 immunohistochemical staining. RESULTS The current study demonstrated that administration of curcumin significantly attenuates spatial memory impairment (P < 0.01) following methamphetamine neurotoxicity. Curcumin caused a significant increase in the levels of superoxide dismutase and glutathione peroxidase (P < 0.05). However, it decreased tumor necrosis factor (TNF-α) (P < 0.05) and malondialdehyde (P < 0.01) levels as compared to the methamphetamine group. Also, curcumin significantly reduced Iba-1 (P < 0. 01), GFAP and caspase-3 positive cells in the hippocampus (P < 0.001). CONCLUSION Curcumin exerted neuroprotective effects on methamphetamine neurotoxicity because of its antioxidant and anti-inflammatory effect.
Collapse
Affiliation(s)
| | - Golamhassan Vaezi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Mehdi Khaksari
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Vida Hojati
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
18
|
Tang Y, Liu J, Wang Y, Yang L, Han B, Zhang Y, Bai Y, Shen L, Li M, Jiang T, Ye Q, Yu X, Huang R, Zhang Z, Xu Y, Yao H. PARP14 inhibits microglial activation via LPAR5 to promote post-stroke functional recovery. Autophagy 2020; 17:2905-2922. [PMID: 33317392 DOI: 10.1080/15548627.2020.1847799] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stroke is a major public health problem leading to high rates of death and disability worldwide, but no effective pharmacological therapy is currently available except for the use of PLAT (plasminogen activator, tissue). Here we show that PARP14 (poly (ADP-ribose) polymerase family, member 14) level was significantly increased in the peri-infarct zone of photothrombotic stroke (PT) mice. Genetic knockdown and pharmacological inhibition of PARP14 aggravated functional impairment and increased infarct volume in PT mice, while overexpression of PARP14 displayed the opposite effects. Furthermore, PARP14 was abundant in microglia, and downregulation of PARP14 increased post-stroke microglial activation, whereas overexpression of PARP14 alleviated microglial activation, possibly through microglial macroautophagy/autophagy modulation. Mechanistically, overexpression of PARP14 suppressed Lpar5 (lysophosphatidic acid receptor 5) gene transcription to inhibit microglial activation post stroke. Taken together, PARP14 is a stroke-induced signal that restricts microglial activation and promotes functional recovery, and can serve as a novel target to develop new therapeutic agents for stroke. Moreover, these findings may be conducive to proper use of various PARP inhibitors.Abbreviations: 3-MA: 3-methyladenine; AIF1/Iba-1: allograft inflammatory factor 1; CNS: central nervous system; CQ: chloroquine; DAPI: 4',6-diamidino-2-phenylindole; DMEM: Dulbecco's modified Eagle's medium; DMSO: dimethyl sulfoxide; ELISA: enzyme-linked immunosorbent assay; FBS: fetal bovine serum; GFAP: glial fibrillary acidic protein; IL1B/IL-1β: interleukin 1 beta; IL6/IL-6: interleukin 6; LPAR5: lysophosphatidic acid receptor 5; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; NOS2/iNOS: nitric oxide synthase 2, inducible; OGD: oxygen glucose deprivation; PAR: polymer of poly (ADP ribose); PARP: poly (ADP-ribose) polymerase family; PBS: phosphate-buffered saline; PLAT/tPA: plasminogen activator, tissue; PT: photothrombotic stroke; qPCR: quantitative polymerase chain reaction; Rap: rapamycin; RBFOX3/NeuN: RNA binding protein, fox-1 homolog (C. elegans) 3; SQSTM1: sequestosome 1; TNF/TNF-α: tumor necrosis factor.
Collapse
Affiliation(s)
- Ying Tang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Jinchang Liu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Yu Wang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Li Yang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Bing Han
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Yuan Zhang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Ying Bai
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Ling Shen
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Mingyue Li
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Teng Jiang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Qingqing Ye
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xiaoyu Yu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Rongrong Huang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yungen Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China.,Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
19
|
Ito-Kureha T, Miyao T, Nishijima S, Suzuki T, Koizumi SI, Villar-Briones A, Takahashi A, Akiyama N, Morita M, Naguro I, Ishikawa H, Ichijo H, Akiyama T, Yamamoto T. The CCR4-NOT deadenylase complex safeguards thymic positive selection by down-regulating aberrant pro-apoptotic gene expression. Nat Commun 2020; 11:6169. [PMID: 33268794 PMCID: PMC7710727 DOI: 10.1038/s41467-020-19975-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 11/05/2020] [Indexed: 12/25/2022] Open
Abstract
A repertoire of T cells with diverse antigen receptors is selected in the thymus. However, detailed mechanisms underlying this thymic positive selection are not clear. Here we show that the CCR4-NOT complex limits expression of specific genes through deadenylation of mRNA poly(A) tails, enabling positive selection. Specifically, the CCR4-NOT complex is up-regulated in thymocytes before initiation of positive selection, where in turn, it inhibits up-regulation of pro-apoptotic Bbc3 and Dab2ip. Elimination of the CCR4-NOT complex permits up-regulation of Bbc3 during a later stage of positive selection, inducing thymocyte apoptosis. In addition, CCR4-NOT elimination up-regulates Dab2ip at an early stage of positive selection. Thus, CCR4-NOT might control thymocyte survival during two-distinct stages of positive selection by suppressing expression levels of pro-apoptotic molecules. Taken together, we propose a link between CCR4-NOT-mediated mRNA decay and T cell selection in the thymus.
Collapse
Affiliation(s)
- Taku Ito-Kureha
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 904-0495, Japan
| | - Takahisa Miyao
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Saori Nishijima
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 904-0495, Japan
| | - Toru Suzuki
- Laboratory for Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Shin-Ichi Koizumi
- Immune Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 904-0495, Japan
| | - Alejandro Villar-Briones
- Instrumental Analysis Section, Research Support Division, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 904-0495, Japan
| | - Akinori Takahashi
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 904-0495, Japan
| | - Nobuko Akiyama
- Laboratory for Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Masahiro Morita
- Department of Molecular Medicine and Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Isao Naguro
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Hiroki Ishikawa
- Immune Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 904-0495, Japan
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Taishin Akiyama
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.
| | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 904-0495, Japan.
- Laboratory for Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.
| |
Collapse
|
20
|
Yi S, Liu YP, Li XY, Yuan XY, Wang Y, Cai Y, Lei YD, Huang L, Zhang ZH. The expression profile and bioinformatics analysis of microRNAs in human bronchial epithelial cells treated by beryllium sulfate. J Appl Toxicol 2020; 41:1275-1285. [PMID: 33197057 DOI: 10.1002/jat.4116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/17/2020] [Accepted: 11/02/2020] [Indexed: 11/08/2022]
Abstract
Beryllium and its compounds are systemic toxicants that mainly accumulate in the lungs. As a regulator of gene expression, microRNAs (miRNAs) were involved in some lung diseases. This study aimed to analyze the levels of some inflammatory cytokine and the differential expressions of miRNAs in human bronchial epithelial cells (16HBE) induced by beryllium sulfate (BeSO4 ) and to further explore the biological functions of differentially expressed miRNAs. The profile of miRNAs in 16HBE cells was detected using the high-throughput sequencing between the control groups (n = 3) and the 150 μmol/L of BeSO4 -treated groups (n = 3). Bioinformatics analysis of differentially expressed miRNAs was performed, including the prediction of target genes, Gene Ontology (GO) analysis, and the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Quantitative real-time polymerase chain reaction (qRT-PCR) was applied to verify some damage-related miRNAs. We found that BeSO4 can increase the levels of some inflammatory cytokine such as interleukin-10 (IL-10), tumor necrosis factor-alpha (TNF-α), interferon-γ (IFN-γ), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2). And BeSO4 altered miRNAs expression of 16HBE cells and a total of 179 differentially expressed miRNAs were identified, including 88 upregulated miRNAs and 91 downregulated miRNAs. The target genes predicted by 28 dysregulated miRNAs were mainly involved in the transcription regulation, signal transduction, MAPK, and VEGF signaling pathway. The qRT-PCR verification results were consistent with the sequencing results. miRNA expression profiling in 16HBE cells exposed to BeSO4 provides new insights into the toxicity mechanism of beryllium exposure.
Collapse
Affiliation(s)
- Shan Yi
- School of Public Health, University of South China, Hengyang, China
| | - Yan-Ping Liu
- School of Public Health, University of South China, Hengyang, China
| | - Xun-Ya Li
- School of Public Health, University of South China, Hengyang, China
| | - Xiao-Yan Yuan
- School of Public Health, University of South China, Hengyang, China
| | - Ye Wang
- School of Public Health, University of South China, Hengyang, China
| | - Ying Cai
- School of Public Health, University of South China, Hengyang, China
| | - Yuan-di Lei
- School of Public Health, University of South China, Hengyang, China
| | - Lian Huang
- School of Public Health, University of South China, Hengyang, China
| | - Zhao-Hui Zhang
- School of Public Health, University of South China, Hengyang, China
| |
Collapse
|
21
|
He C, Bai Y, Wang Z, Fan D, Wang Q, Liu X, Zhang H, Zhang H, Zhang Z, Yao H, Xie C. Identification of microRNA-9 linking the effects of childhood maltreatment on depression using amygdala connectivity. Neuroimage 2020; 224:117428. [PMID: 33038536 DOI: 10.1016/j.neuroimage.2020.117428] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 01/20/2023] Open
Abstract
Childhood maltreatment (CM) is regarded as an important risk factor for major depressive disorder (MDD). However, the neural links corresponding to the process of early CM experience producing brain alterations and then leading to depression later remain unclear. To explore the neural basis of the effects of CM on MDD and the potential role of microRNA-9 (miR-9) in these processes, we recruited 40 unmedicated MDD patients and 34 healthy controls (HCs) to complete resting-state fMRI scans and peripheral blood miR-9 tests. The neural substrates of CM, miR-9, and depression, as well as their interactive effects on intrinsic amygdala functional connectivity (AFC) networks were investigated in MDD patients. Two-step mediation analysis was separately employed to explore whether AFC strength mediates the association among CM severity, miR-9 levels, and depression. A support vector classifier (SVC) model of machine learning was used to distinguish MDD patients from HCs. MDD patients showed higher miR-9 levels that were negatively correlated with CM scores and depressive severity. Overlapping effects of CM, miR-9, and depressive severity on bilateral AFC networks in MDD patients were primarily located in the prefrontal-striatum pathway and limbic system. The connection of amygdala to prefrontal-limbic circuits could mediate the effects of CM severity on the miR-9 levels, as well as the impacts of miR-9 levels on the severity of depression in MDD patients. Furthermore, the SVC model, which integrated miR-9 levels, CM severity, and AFC strength in prefrontal-limbic regions, had good power in differentiating MDD patients from HCs (accuracy 85.1%). MiR-9 may play a crucial role in the process of CM experience-produced brain changes targeting prefrontal-limbic regions and that subsequently leads to depression. The present neuroimaging-epigenetic results provide new insight into our understanding of MDD pathophysiology.
Collapse
Affiliation(s)
- Cancan He
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Ying Bai
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Zan Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China; Institute of Neuropsychiatry, Affiliated ZhongDa Hospital, Southeast University, Nanjing, Jiangsu 210009, China
| | - Dandan Fan
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Qing Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Xinyi Liu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Haisan Zhang
- Department of Radiology, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China; Xinxiang Key Laboratory of Multimodal Brain Imaging, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Hongxing Zhang
- Department of Psychiatry, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China; Psychology School of Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China; Institute of Neuropsychiatry, Affiliated ZhongDa Hospital, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China; Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210096, China.
| | - Chunming Xie
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China; Institute of Neuropsychiatry, Affiliated ZhongDa Hospital, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
22
|
Li J, Sun Q, Zhu S, Xi K, Shi Q, Pang K, Liu X, Li M, Zhang Y, Sun J. Knockdown of circHomer1 ameliorates METH-induced neuronal injury through inhibiting Bbc3 expression. Neurosci Lett 2020; 732:135050. [PMID: 32450188 DOI: 10.1016/j.neulet.2020.135050] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/25/2020] [Accepted: 05/10/2020] [Indexed: 12/19/2022]
Abstract
Current studies have illustrated that circular RNAs (circRNAs) are a vital part of non-coding RNA (ncRNAs) species and highly abundant and dynamically expressed in brain. However, the exact mechanisms by which circRNAs modulate methamphetamine (METH)-induced neuronal damage still remain largely unexplored. Consistent with our previous study, the expression of circHomer1 was significantly up-regulated after METH treatment in HT-22 cells. We confirmed its loop structure by detection of its back-splice junction with qRT-PCR product via sequence. Moreover, circHomer1 was resistant against RNase R digestion compared with its linear mRNA Homer1. Inhibition of circHomer1 expression indeed alleviated METH-induced neurotoxicity, with lower apoptosis rate via flow cytometry and cleaved Caspase3 protein level. Furthermore, we speculated that Bbc3 functioned as a target of circHomer1 based on computational algorithm, and knockdown of circHomer1 actually reduced Bbc3 expression at the mRNA and protein level. Besides, suppression of Bbc3 decreased the reactive oxygen species (ROS) level and radio of PI-positive cells. Furthermore, we analyzed the correlation in pairs among circHomer1, Bbc3 and behaviors in well-developed METH-addicted models using Pearson's correlation coefficient, which implied an important role of circHomer1 and Bbc3 in addictive behaviors. In all, we for the first time identified a novel circRNA, circHomer1 and our results suggested that circHomer1 regulated METH-induced lethal process by suppressing the Bbc3 expression.
Collapse
Affiliation(s)
- Junwei Li
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Qiyun Sun
- Department of Orthopedics, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, 277100, China
| | - Shaowei Zhu
- Department of Neurology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Kaiyan Xi
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Qingqing Shi
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Kunkun Pang
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Xiaoyu Liu
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Meng Li
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Yue Zhang
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Jinhao Sun
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
23
|
Yu S, Yu M, Bu Z, He P, Feng J. FKBP5 Exacerbates Impairments in Cerebral Ischemic Stroke by Inducing Autophagy via the AKT/FOXO3 Pathway. Front Cell Neurosci 2020; 14:193. [PMID: 32760250 PMCID: PMC7374263 DOI: 10.3389/fncel.2020.00193] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/04/2020] [Indexed: 01/01/2023] Open
Abstract
Cerebral ischemic stroke is regarded as one of the most serious diseases in the human central nervous system. The secondary ischemia and reperfusion (I/R) injury increased the difficulty of treatment. Moreover, the latent molecular regulating mechanism in I/R injury is still unclear. Based on our previous clinical study, we discovered that FK506 binding protein 5 (FKBP5) is significantly upregulated in patients, who suffered acute ischemic stroke (AIS), with high diagnostic value. Levels of FKBP5 were positively correlated with patients’ neurological impairments. Furthermore, a transient middle cerebral artery occlusion (tMCAO) model of mice was used to confirm that FKBP5 expression in plasma could reflect its relative level in brain tissue. Thus, we hypothesized that FKBP5 participated in the regulation of cerebral I/R injury. In order to explore the possible roles FKBP5 acted, the oxygen and glucose deprivation and reoxygenation (OGD/R) model was established to mimic I/R injury in vitro. FKBP5 expressing levels were changed by plasmid stable transfection. The altered expression of FKBP5 influenced cell viability and autophagy after OGD/R injury notably. Besides, AKT/FOXO3 cascade was involved in the FKBP5-regulating process. In the present study, FKBP5 was verified upregulated in cerebral I/R injury, related to the severity of ischemia and reperfusion injury. Additionally, our analyses revealed that FKBP5 regulates autophagy induced by OGD/R via the downstream AKT/FOXO3 signaling pathway. Our findings provide a novel biomarker for the early diagnosis of ischemic stroke and a potential strategy for treatment.
Collapse
Affiliation(s)
- Shijia Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mingjun Yu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhongqi Bu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Pingping He
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
24
|
Brites D. Regulatory function of microRNAs in microglia. Glia 2020; 68:1631-1642. [PMID: 32463968 DOI: 10.1002/glia.23846] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/14/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022]
Abstract
Microglia are CNS-resident cells involved in immune surveillance and maintenance of intercellular homeostasis, while also contributing to neurologic pathologies. MicroRNAs (miRNAs) are a class of small (~22 nucleotides) single-stranded noncoding RNAs that participate in gene regulation at the post-transcriptional level. miRNAs typically bind to the untranslated region (3' UTR) of RNAs. It has been shown that miRNAs are important players in controlling inflammation and that their abnormal expression is linked to cancer and ageing, and to the onset and progression of neurodegenerative disease. Furthermore, miRNAs participate in intercellular trafficking. Thus, miRNAs are released from cells in a free form, bound to proteins or packaged within extracellular vesicles (EVs), exerting paracrine and long distance signaling. In this review, recent findings on the role of miRNAs as drivers of microglia phenotypic changes and their cotribution in neurological disease are addressed. MAIN POINTS: miRNAs have a key role in microglia function/dysfunction, polarization, and restoration. Microglia are both a source and recipient of extracellular vesicles (EVs) containing miRNAs. Extracellular miRNAs may be found as soluble (free and EV cargo) and protein complexes.
Collapse
Affiliation(s)
- Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
25
|
Thangaraj A, Sil S, Tripathi A, Chivero ET, Periyasamy P, Buch S. Targeting endoplasmic reticulum stress and autophagy as therapeutic approaches for neurological diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 350:285-325. [DOI: 10.1016/bs.ircmb.2019.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
CircDYM ameliorates depressive-like behavior by targeting miR-9 to regulate microglial activation via HSP90 ubiquitination. Mol Psychiatry 2020; 25:1175-1190. [PMID: 30413800 PMCID: PMC7244405 DOI: 10.1038/s41380-018-0285-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 08/06/2018] [Accepted: 09/06/2018] [Indexed: 11/23/2022]
Abstract
Circular RNAs (circRNAs), highly expressed in the central nervous system, are involved in various regulatory processes and implicated in some pathophysiology. However, the potential role of circRNAs in psychiatric diseases, particularly major depressive disorder (MDD), remains largely unknown. Here, we demonstrated that circular RNA DYM (circDYM) levels were significantly decreased both in the peripheral blood of patients with MDD and in the two depressive-like mouse models: the chronic unpredictable stress (CUS) and lipopolysaccharide (LPS) models. Restoration of circDYM expression significantly attenuated depressive-like behavior and inhibited microglial activation induced by CUS or LPS treatment. Further examination indicated that circDYM functions as an endogenous microRNA-9 (miR-9) sponge to inhibit miR-9 activity, which results in a downstream increase of target-HECT domain E3 ubiquitin protein ligase 1 (HECTD1) expression, an increase of HSP90 ubiquitination, and a consequent decrease of microglial activation. Taken together, the results of our study demonstrate the involvement of circDYM and its coupling mechanism in depression, providing translational evidence that circDYM may be a novel therapeutic target for depression.
Collapse
|
27
|
Tp47 induces cell death involving autophagy and mTOR in human microglial HMO6 cells. Int Immunopharmacol 2019; 74:105566. [DOI: 10.1016/j.intimp.2019.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/31/2019] [Accepted: 04/04/2019] [Indexed: 01/24/2023]
|
28
|
Chen X, Lu J, Zhao X, Chen C, Qiao D, Wang H, Yue X. Role of C/EBP-β in Methamphetamine-Mediated Microglial Apoptosis. Front Cell Neurosci 2019; 13:366. [PMID: 31496936 PMCID: PMC6712175 DOI: 10.3389/fncel.2019.00366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/29/2019] [Indexed: 12/14/2022] Open
Abstract
Methamphetamine (MA) is a widely abused psychoactive drug that primarily damages the nervous system. However, the involvement of MA in the survival of microglia remains poorly understood. CCAAT-enhancer binding protein (C/EBP-β) is a transcription factor and an important regulator of cell apoptosis. Lipocalin2 (lcn2) is a known apoptosis inducer and is involved in many cell death processes. We hypothesized that C/EBP-β is involved in MA-induced lcn2-mediated microglial apoptosis. To test this hypothesis, we measured the protein expression of C/EBP-β after MA treatment and evaluated the effects of silencing C/EBP-β or lcn2 on MA-induced apoptosis in BV-2 cells and the mouse striatum after intrastriatal MA injection. MA exposure increased the expression of C/EBP-β and stimulated the lcn2-mediated modulation of apoptosis. Moreover, silencing the C/EBP-β-dependent lcn2 upregulation reversed the MA-induced microglial apoptosis. The in vivo relevance of these findings was confirmed in mouse models, which demonstrated that the microinjection of anti-C/EBP-β into the striatum ameliorated the MA-induced decrease survival of microglia. These findings provide a new insight regarding the specific contributions of C/EBP-β-lcn2 to microglial survival in the context of MA abuse.
Collapse
Affiliation(s)
- Xuebing Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Jiancong Lu
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xu Zhao
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Chuanxiang Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Dongfang Qiao
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xia Yue
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
29
|
Circular RNA TLK1 Aggravates Neuronal Injury and Neurological Deficits after Ischemic Stroke via miR-335-3p/TIPARP. J Neurosci 2019; 39:7369-7393. [PMID: 31311824 DOI: 10.1523/jneurosci.0299-19.2019] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 06/30/2019] [Accepted: 07/06/2019] [Indexed: 02/06/2023] Open
Abstract
Circular RNAs (circRNAs) are expressed at high levels in the brain and are involved in various CNS diseases. However, the potential role of circRNAs in ischemic stroke-associated neuronal injury remains largely unknown. Here, we investigated the important functions of circRNA TLK1 (circTLK1) in this process. The levels of circTLK1 were significantly increased in brain tissues in a mouse model of focal cerebral ischemia and reperfusion. Knockdown of circTLK1 significantly decreased infarct volumes, attenuated neuronal injury, and improved neurological deficits. Furthermore, circTLK1 functioned as an endogenous miR-335-3p sponge to inhibit miR-335-3p activity, resulting in the increase of 2,3,7,8-tetrachlorodibenzo-p-dioxin-inducible poly (ADP-ribose) polymerase expression and a subsequent exacerbation of neuronal injury. Clinical studies confirmed increased levels of circTLK1 in the plasma of patients with acute ischemic stroke (59 males and 12 females). Our findings reveal a detrimental role of circTLK1 in ischemic brain injury.SIGNIFICANCE STATEMENT The extent of neuronal injury after brain ischemia is a primary factor determining stroke outcomes. However, the molecular switches that control the death of ischemic neurons are poorly understood. While our previous studies indicated the involvement of circRNAs in ischemic stroke, the potential role of circRNAs in neuronal injury remains largely unknown. The levels of circTLK1 were significantly increased in the brain tissue and plasma isolated from animal models of ischemic stroke and patients. Knockdown of circTLK1 significantly decreased infarct volumes, attenuated neuronal injury, and improved subsequent long-term neurological deficits. To our knowledge, these results provide the first definitive evidence that circTLK1 is detrimental in ischemic stroke.
Collapse
|
30
|
Lu S, Yang X, Wang C, Chen S, Lu S, Yan W, Xiong K, Liu F, Yan J. Current status and potential role of circular RNAs in neurological disorders. J Neurochem 2019; 150:237-248. [PMID: 31099046 DOI: 10.1111/jnc.14724] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/23/2019] [Accepted: 05/13/2019] [Indexed: 01/01/2023]
Abstract
Given the importance of non-coding RNAs in modulating normal brain functions and their implications in the treatment of neurological disorders, non-coding RNA-based diagnostic and therapeutic strategies have shown great clinical potential. Circular RNAs (circRNAs) have emerged as potentially important players in this field. Recent studies have indicated that circRNAs might play vital roles in Alzheimer's disease, Parkinson's disease, ischemic brain injury, and neurotoxicity. However, the mechanisms of action of circRNAs have not been fully characterized. We aimed to review recent advances in circRNA research in the brain to provide new insights on the roles of circRNAs in neurological disorders.
Collapse
Affiliation(s)
- Shanshan Lu
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Histology and Embryology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xue Yang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Chudong Wang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Siqi Chen
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Shuang Lu
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Weitao Yan
- Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Kun Xiong
- Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Fengxia Liu
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
31
|
Guo Y, Hong W, Wang X, Zhang P, Körner H, Tu J, Wei W. MicroRNAs in Microglia: How do MicroRNAs Affect Activation, Inflammation, Polarization of Microglia and Mediate the Interaction Between Microglia and Glioma? Front Mol Neurosci 2019; 12:125. [PMID: 31133802 PMCID: PMC6522842 DOI: 10.3389/fnmol.2019.00125] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/26/2019] [Indexed: 12/31/2022] Open
Abstract
The essential roles of microglia in maintaining homeostasis in the healthy brain and contributing to neuropathology are well documented. Emerging evidence suggests that epigenetic modulation regulates microglial behavior in both physiological and pathological conditions. MicroRNAs (miRNAs) are short, non-coding epigenetic regulators that repress target gene expression mostly via binding to 3'-untranslated region (3'-UTR) of mRNA in a Dicer-dependent manner. Dysregulation of certain miRNAs can contribute to microglial hyper-activation, persistent neuroinflammation, and abnormal macrophage polarization in the brain. These abnormal conditions can support the pathogenesis of neurological disorders such as glioma, Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), stroke, ischemia, and spinal cord injury (SCI). However, the roles of miRNAs in microglia in health and neurological disease have not been systematically summarized. This review will first report the role of Dicer, a key endoribonulease that is responsible for most miRNA biogenesis in microglia. Second, we will focus on recent research about the function of miRNAs in activation, inflammation and polarization of microglia, respectively. In addition, potential crosstalk between microglia and glioma cells via miRNAs will be discussed in this part. Finally, the role of two essential miRNAs, miR-124, and miR-155, in microglia will be highlighted.
Collapse
Affiliation(s)
- Yawei Guo
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Wenming Hong
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xinming Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Pengying Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Heinrich Körner
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Jiajie Tu
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| |
Collapse
|
32
|
Catale C, Bussone S, Lo Iacono L, Carola V. Microglial alterations induced by psychoactive drugs: A possible mechanism in substance use disorder? Semin Cell Dev Biol 2019; 94:164-175. [PMID: 31004753 DOI: 10.1016/j.semcdb.2019.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/19/2019] [Accepted: 03/29/2019] [Indexed: 12/11/2022]
Abstract
Recently, the xenobiotic hypothesis has implicated the immune system in targeting substances of abuse as foreign molecules and stimulating inflammatory responses. Microglial cells are the resident immune cells of the central nervous system and function in homeostatic surveillance. Microglial changes that are induced by exposure to substances of abuse appear to mediate in part the establishment of addiction and the persistence of drug-mediated biological and behavioral changes. In this context, interest in the study of drug-microglia interactions has increased recently. This review summarizes the most recent preclinical rodent and clinical studies on the interaction between microglia and various classes of drugs of abuse, such as ethanol, psychostimulants, and opioids. The principal biological mechanisms of the communication between substances of abuse and microglia will be described to consider putative mechanisms of the establishment of drug addiction and future potential targets for treating substance use disorder.
Collapse
Affiliation(s)
- Clarissa Catale
- Department of Psychology, University of Rome "La Sapienza", Via dei Marsi, 78, 00185 Rome, Italy
| | - Silvia Bussone
- Department of Dynamic and Clinical Psychology, University of Rome "La Sapienza", Via degli Apuli 1, 00185 Rome, Italy
| | - Luisa Lo Iacono
- Department of Psychology, University of Rome "La Sapienza", Via dei Marsi, 78, 00185 Rome, Italy; IRCCS Santa Lucia Foundation, Via Fosso di Fiorano 64, 00143 Rome, Italy
| | - Valeria Carola
- Department of Dynamic and Clinical Psychology, University of Rome "La Sapienza", Via degli Apuli 1, 00185 Rome, Italy; IRCCS Santa Lucia Foundation, Via Fosso di Fiorano 64, 00143 Rome, Italy.
| |
Collapse
|
33
|
Liu HQ, An YW, Hu AZ, Li MH, Wu JL, Liu L, Shi Y, Cui GH, Chen Y. Critical Roles of the PI3K-Akt-mTOR Signaling Pathway in Apoptosis and Autophagy of Astrocytes Induced by Methamphetamine. OPEN CHEM 2019. [DOI: 10.1515/chem-2019-0015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AbstractThis study aimed to reveal potential roles of the phosphatidylinositol 3 kinase (PI3K)-protein kinase B (Akt)-mammalian target of rapamycin (mTOR) signaling pathway in apoptosis and autophagy of astrocytes induced by methamphetamine (METH). A Cell Counting Kit-8 (CCK-8) was used to determine the reduction in proliferation of U-118 MG cells induced by METH. Hoechst 33258 and flow cytometry were used to observe the astrocytes. Western blot analysis was performed to evaluate protein expression and phosphorylation levels. METH inhibited the proliferation of U-118 MG cells and induced apoptosis and autophagy. Western blot analysis showed that the ratio of LC3-II/I was increased, whereas the expression of Bcl-2 was decreased. The phosphorylation cascade of kinases in the PI3K-Akt-mTOR signaling pathway was significantly inhibited by METH exposure, as were proteins downstream of mTORC1, such as p70s6k, rps6, 4EBP1 and eIF4E. METH inhibited proliferation of U-118 MG cells and induced apoptosis and autophagy. The PI3K-Akt-mTOR signaling pathway likely plays a critical role in these effects.
Collapse
Affiliation(s)
- Han-Qing Liu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen City, Guangdong Province, China, 518036
| | - Ya-Wen An
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen City, Guangdong Province, China, 518055
| | - A-Zhen Hu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen City, Guangdong Province, China, 518036
| | - Ming-Hua Li
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen City, Guangdong Province, China, 518036
| | - Jue-Lian Wu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen City, Guangdong Province, China, 518036
| | - Li Liu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen City, Guangdong Province, China, 518036
| | - Yu Shi
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen City, Guangdong Province, China, 518036
| | - Guang-Hui Cui
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen City, Guangdong Province, China, 518036
| | - Yun Chen
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen City, Guangdong Province, China, 518036
| |
Collapse
|
34
|
Du L, Shen K, Bai Y, Chao J, Hu G, Zhang Y, Yao H. Involvement of NLRP3 inflammasome in methamphetamine-induced microglial activation through miR-143/PUMA axis. Toxicol Lett 2019; 301:53-63. [PMID: 30394308 DOI: 10.1016/j.toxlet.2018.10.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 10/02/2018] [Accepted: 10/22/2018] [Indexed: 12/11/2022]
Abstract
Nod-like Receptor Protein 3 (NLRP3) inflammasome activation is known to lead to microglia-mediated neuroinflammation. Methamphetamine is known to induce microglial activation. However, whether NLRP3 inflammasome activation contributes to the microglial activation induced by methamphetamine remains elusive. P53-up-regulated modulator of apoptosis (PUMA) is a known apoptosis inducer; however, their role in microglial activation remains poorly understood. Methamphetamine treatment induced NLRP3 inflammasome activation as well microglial activation in animal model. Intriguingly, downregulation of PUMA significantly inhibited the activation of microglia. Methamphetamine treatment increased the expression of PUMA at protein level but not mRNA level. Further study indicated that PUMA expression was regulated at post-transcriptional level by miR-143, which was decreased in methamphetamine-treated cells via the negative transcription factor nuclear factor-kappa B1 (NF-κB1). Using gain- and loss-of-function approaches, we identified a unique role of miR-143/PUMA in mediating microglial activation via regulation of NLRP3 inflammasome activation. These findings provide new insight regarding the specific contributions of the miR-143/PUMA pathway to NLRP3 inflammasome activation in the context of drug abuse.
Collapse
Affiliation(s)
- Longfei Du
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Kai Shen
- Department of Pharmacology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Ying Bai
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Jie Chao
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yuan Zhang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China.
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China; Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210096, Jiangsu, China.
| |
Collapse
|
35
|
Han B, Zhang Y, Zhang Y, Bai Y, Chen X, Huang R, Wu F, Leng S, Chao J, Zhang JH, Hu G, Yao H. Novel insight into circular RNA HECTD1 in astrocyte activation via autophagy by targeting MIR142-TIPARP: implications for cerebral ischemic stroke. Autophagy 2018; 14:1164-1184. [PMID: 29938598 DOI: 10.1080/15548627.2018.1458173] [Citation(s) in RCA: 276] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Circular RNAs (circRNAs) are highly expressed in the central nervous system and are involved in the regulation of physiological and pathophysiological processes. However, the potential role of circRNAs in stroke remains largely unknown. Here, using a circRNA microarray, we showed that circular RNA Hectd1 (circHectd1) levels were significantly increased in ischemic brain tissues in transient middle cerebral artery occlusion (tMCAO) mouse stroke models and further validated this finding in plasma samples from acute ischemic stroke (AIS) patients. Knockdown of circHectd1 expression significantly decreased infarct areas, attenuated neuronal deficits, and ameliorated astrocyte activation in tMCAO mice. Mechanistically, circHECTD1 functions as an endogenous MIR142 (microRNA 142) sponge to inhibit MIR142 activity, resulting in the inhibition of TIPARP (TCDD inducible poly[ADP-ribose] polymerase) expression with subsequent inhibition of astrocyte activation via macroautophagy/autophagy. Taken together, the results of our study indicate that circHECTD1 and its coupling mechanism are involved in cerebral ischemia, thus providing translational evidence that circHECTD1 can serve as a novel biomarker of and therapeutic target for stroke. ABBREVIATIONS 3-MA: 3-methyladenine; ACTB: actin beta; AIS: acute ischemic stroke; AS: primary mouse astrocytes; BECN1: beclin 1, autophagy related; BMI: body mass index; circHECTD1: circRNA HECTD1; circRNAs: circular RNAs; CBF: cerebral blood flow; Con: control; DAPI: 4',6-diamidino-2-phenylindole; ECA: external carotid artery; FISH: fluorescence in situ hybridization; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; Gdna: genomic DNA; GFAP: glial fibrillary acidic protein; GO: gene ontology; HDL: high-density lipoprotein; IOD: integrated optical density; LDL: low-density lipoprotein; LPA: lipoprotein(a); MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; MIR142: microRNA 142; mNSS: modified neurological severity scores; MRI: magnetic resonance imaging; NIHSS: National Institute of Health Stoke Scale; OGD-R: oxygen glucose deprivation-reperfusion; PCR: polymerase chain reaction; PFA: paraformaldehyde; SQSTM1: sequestosome 1; TIPARP: TCDD inducible poly(ADP-ribose) polymerase; tMCAO: transient middle cerebral artery occlusion; TTC: 2,3,5-triphenyltetrazolium chloride; UTR: untranslated region; WT: wild type.
Collapse
Affiliation(s)
- Bing Han
- a Department of Pharmacology , School of Medicine, Southeast University , Nanjing , Jiangsu , China
| | - Yuan Zhang
- a Department of Pharmacology , School of Medicine, Southeast University , Nanjing , Jiangsu , China
| | - Yanhong Zhang
- a Department of Pharmacology , School of Medicine, Southeast University , Nanjing , Jiangsu , China
| | - Ying Bai
- a Department of Pharmacology , School of Medicine, Southeast University , Nanjing , Jiangsu , China
| | - Xufeng Chen
- b Department of Emergency , Jiangsu Province Hospital and The First Affiliated Hospital of Nanjing Medical University , Nanjing , Jiangsu , China
| | - Rongrong Huang
- a Department of Pharmacology , School of Medicine, Southeast University , Nanjing , Jiangsu , China
| | - Fangfang Wu
- a Department of Pharmacology , School of Medicine, Southeast University , Nanjing , Jiangsu , China
| | - Shuo Leng
- c Department of Radiology , School of Medicine, Southeast University , Nanjing , Jiangsu , China
| | - Jie Chao
- d Department of Physiology , School of Medicine, Southeast University , Nanjing , Jiangsu , China
| | - John H Zhang
- e Department of Physiology and Pharmacology , School of Medicine, Loma Linda University , Loma Linda , California , USA
| | - Gang Hu
- f Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology , Nanjing Medical University , Nanjing , Jiangsu , China
| | - Honghong Yao
- a Department of Pharmacology , School of Medicine, Southeast University , Nanjing , Jiangsu , China.,g Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease , Southeast University , Nanjing , Jiangsu , China
| |
Collapse
|
36
|
Yang X, Wang Y, Li Q, Zhong Y, Chen L, Du Y, He J, Liao L, Xiong K, Yi CX, Yan J. The Main Molecular Mechanisms Underlying Methamphetamine- Induced Neurotoxicity and Implications for Pharmacological Treatment. Front Mol Neurosci 2018; 11:186. [PMID: 29915529 PMCID: PMC5994595 DOI: 10.3389/fnmol.2018.00186] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/14/2018] [Indexed: 01/07/2023] Open
Abstract
Methamphetamine (METH) is a popular new-type psychostimulant drug with complicated neurotoxicity. In spite of mounting evidence on METH-induced damage of neural cell, the accurate mechanism of toxic effect of the drug on central nervous system (CNS) has not yet been completely deciphered. Besides, effective treatment strategies toward METH neurotoxicity remain scarce and more efficacious drugs are to be developed. In this review, we summarize cellular and molecular bases that might contribute to METH-elicited neurotoxicity, which mainly include oxidative stress, excitotoxicity, and neuroinflammation. We also discuss some drugs that protect neural cells suffering from METH-induced neurotoxic consequences. We hope more in-depth investigations of exact details that how METH produces toxicity in CNS could be carried out in future and the development of new drugs as natural compounds and immunotherapies, including clinic trials, are expected.
Collapse
Affiliation(s)
- Xue Yang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yong Wang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Qiyan Li
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yaxian Zhong
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Liangpei Chen
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yajun Du
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Jing He
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Lvshuang Liao
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
37
|
Ma W, Ding F, Wang X, Huang Q, Zhang L, Bi C, Hua B, Yuan Y, Han Z, Jin M, Liu T, Yu Y, Cai B, Du Z. By Targeting Atg7 MicroRNA-143 Mediates Oxidative Stress-Induced Autophagy of c-Kit + Mouse Cardiac Progenitor Cells. EBioMedicine 2018; 32:182-191. [PMID: 29858017 PMCID: PMC6021267 DOI: 10.1016/j.ebiom.2018.05.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/01/2018] [Accepted: 05/16/2018] [Indexed: 12/19/2022] Open
Abstract
Therapeutic efficiency of cardiac progenitor cells (CPCs) transplantation is limited by its low survival and retention in infarcted myocardium. Autophagy plays a critical role in regulating cell death and apoptosis, but the role of microRNAs (miRNAs) in oxidative stress-induced autophagy of CPCs remains unclear. This study aimed to explore if miRNAs mediate autophagy of c-kit+ CPCs. We found that the silencing of miR-143 promoted the autophagy of c-kit+ CPCs in response to H2O2, and the protective effect of miR-143 inhibitor was abrogated by autophagy inhibitor 3-methyladenine (3-MA). Furthermore, autophagy-related gene 7 (Atg7) was identified as the target gene of miR-143 by dual luciferase reporter assays. In vivo, after transfection with miR-143 inhibitor, c-kit+ CPCs from green fluorescent protein transgenic mice were more observed in infarcted mouse hearts. Moreover, transplantation of c-kit+ CPCs with miR-143 inhibitor improved cardiac function after myocardial infarction. Take together, our study demonstrated that miR-143 mediates oxidative stress-induced autophagy to enhance the survival of c-kit+ CPCs by targeting Atg7, which will provide a complementary approach for improving CPC-based heart repair.
Collapse
Affiliation(s)
- Wenya Ma
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China; Institute of Clinical Pharmacy, Harbin Medical University, Harbin, China
| | - Fengzhi Ding
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xiuxiu Wang
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Qi Huang
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Lai Zhang
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Chongwei Bi
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Bingjie Hua
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ye Yuan
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhenbo Han
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Mengyu Jin
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Tianyi Liu
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ying Yu
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Benzhi Cai
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China; Institute of Clinical Pharmacy, Harbin Medical University, Harbin, China.
| | - Zhimin Du
- Department of Pharmacy of The Second Affiliated Hospital, Harbin Medical University (Heilongjiang Provincial Key Laboratory of Drug Research, Harbin Medical University), Harbin, China; Department of Pharmacology (Key Laboratoryof Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China; Institute of Clinical Pharmacy, Harbin Medical University, Harbin, China.
| |
Collapse
|
38
|
Wang Y, Tang M. Dysfunction of various organelles provokes multiple cell death after quantum dot exposure. Int J Nanomedicine 2018; 13:2729-2742. [PMID: 29765216 PMCID: PMC5944465 DOI: 10.2147/ijn.s157135] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Quantum dots (QDs) are different from the materials with the micrometer scale. Owing to the superiority in fluorescence and optical stability, QDs act as possible diagnostic and therapeutic tools for application in biomedical field. However, potential threats of QDs to human health hamper their wide utilization in life sciences. It has been reported that oxidative stress and inflammation are involved in toxicity caused by QDs. Recently, accumulating research unveiled that disturbance of subcellular structures plays a magnificent role in cytotoxicity of QDs. Diverse organelles would collapse during QD treatment, including DNA damage, endoplasmic reticulum stress, mitochondrial dysfunction and lysosomal rupture. Different forms of cellular end points on the basis of recent research have been concluded. Apart from apoptosis and autophagy, a new form of cell death termed pyroptosis, which is finely orchestrated by inflammasome complex and gasdermin family with secretion of interleukin-1 beta and interleukin-18, was also summarized. Finally, several potential cellular signaling pathways were also listed. Activation of Toll-like receptor-4/myeloid differentiation primary response 88, nuclear factor kappa-light-chain-enhancer of activated B cells and NACHT, LRR and PYD domains-containing protein 3 inflammasome pathways by QD exposure is associated with regulation of cellular processes. With the development of QDs, toxicity evaluation is far behind its development, where specific mechanisms of toxic effects are not clearly defined. Further studies concerned with this promising area are urgently required.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Meng Tang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
39
|
Yang L, Han B, Zhang Y, Bai Y, Chao J, Hu G, Yao H. Engagement of circular RNA HECW2 in the nonautophagic role of ATG5 implicated in the endothelial-mesenchymal transition. Autophagy 2018; 14:404-418. [PMID: 29260931 DOI: 10.1080/15548627.2017.1414755] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Endothelial-mesenchymal transition (EndoMT) is associated with damage to blood-brain barrier (BBB) integrity. Circular RNAs (circRNAs) are highly expressed in the brain and are involved in brain diseases; however, whether circRNAs regulate the EndoMT in the brain remains unknown. Our study demonstrated that circHECW2 regulated the EndoMT by directly binding to MIR30D, a significantly downregulated miRNA from miRNA profiling, which subsequently caused an increased expression of ATG5. These findings shed new light on the understanding of the noncanonical role of ATG5 in the EndoMT induced by methamphetamine (Meth) or lipopolysaccharide (LPS). The in vivo relevance was confirmed as microinjection of circHecw2 siRNA lentivirus into the mouse hippocampus suppressed the EndoMT induced by LPS. These findings provide novel insights regarding the contribution of circHECW2 to the nonautophagic role of ATG5 in the EndoMT process in the context of drug abuse and the broad range of neuroinflammatory disorders.
Collapse
Affiliation(s)
- Li Yang
- a Department of Pharmacology, School of Medicine , Southeast University , Nanjing , Jiangsu , China
| | - Bing Han
- a Department of Pharmacology, School of Medicine , Southeast University , Nanjing , Jiangsu , China
| | - Yuan Zhang
- a Department of Pharmacology, School of Medicine , Southeast University , Nanjing , Jiangsu , China
| | - Ying Bai
- a Department of Pharmacology, School of Medicine , Southeast University , Nanjing , Jiangsu , China
| | - Jie Chao
- b Department of Physiology, School of Medicine , Southeast University , Nanjing , Jiangsu , China
| | - Gang Hu
- c Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology , Nanjing Medical University , Nanjing , Jiangsu , China
| | - Honghong Yao
- a Department of Pharmacology, School of Medicine , Southeast University , Nanjing , Jiangsu , China.,d Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease , Southeast University , Nanjing , Jiangsu , China
| |
Collapse
|
40
|
Bai Y, Zhang Y, Han B, Yang L, Chen X, Huang R, Wu F, Chao J, Liu P, Hu G, Zhang JH, Yao H. Circular RNA DLGAP4 Ameliorates Ischemic Stroke Outcomes by Targeting miR-143 to Regulate Endothelial-Mesenchymal Transition Associated with Blood-Brain Barrier Integrity. J Neurosci 2018; 38:32-50. [PMID: 29114076 PMCID: PMC6705810 DOI: 10.1523/jneurosci.1348-17.2017] [Citation(s) in RCA: 296] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 10/24/2017] [Accepted: 10/29/2017] [Indexed: 01/01/2023] Open
Abstract
Circular RNAs (circRNAs) are highly expressed in the CNS and regulate physiological and pathophysiological processes. However, the potential role of circRNAs in stroke remains largely unknown. Here, we show that the circRNA DLGAP4 (circDLGAP4) functions as an endogenous microRNA-143 (miR-143) sponge to inhibit miR-143 activity, resulting in the inhibition of homologous to the E6-AP C-terminal domain E3 ubiquitin protein ligase 1 expression. circDLGAP4 levels were significantly decreased in the plasma of acute ischemic stroke patients (13 females and 13 males) and in a mouse stroke model. Upregulation of circDLGAP4 expression significantly attenuated neurological deficits and decreased infarct areas and blood-brain barrier damage in the transient middle cerebral artery occlusion mouse stroke model. Endothelial-mesenchymal transition contributes to blood-brain barrier disruption and circDLGAP4 overexpression significantly inhibited endothelial-mesenchymal transition by regulating tight junction protein and mesenchymal cell marker expression. Together, the results of our study are illustrative of the involvement of circDLGAP4 and its coupling mechanism in cerebral ischemia, providing translational evidence that circDLGAP4 serves as a novel therapeutic target for acute cerebrovascular protection.SIGNIFICANCE STATEMENT Circular RNAs (circRNAs) are involved in the regulation of physiological and pathophysiological processes. However, whether circRNAs are involved in ischemic injury, particularly cerebrovascular disorders, remains largely unknown. Here, we demonstrate a critical role for circular RNA DLGAP4 (circDLGAP4), a novel circular RNA originally identified as a sponge for microRNA-143 (miR-143), in ischemic stroke outcomes. Overexpression of circDLGAP4 significantly attenuated neurological deficits and decreased infarct areas and blood-brain barrier damage in the transient middle cerebral artery occlusion mouse stroke model. To our knowledge, this is the first report describing the efficacy of circRNA injection in an ischemic stroke model. Our investigation suggests that circDLGAP4 may serve as a novel therapeutic target for acute ischemic injury.
Collapse
Affiliation(s)
- Ying Bai
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yuan Zhang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Bing Han
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Li Yang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xufeng Chen
- Emergency Department, Jiangsu Province Hospital, Nanjing 210029, China
| | - Rongrong Huang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Fangfang Wu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jie Chao
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Pei Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Southeast University, Nanjing 210009, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China
| | - John H Zhang
- Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, California 92354, and
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China,
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| |
Collapse
|
41
|
Zhu J, Yu W, Liu B, Wang Y, Shao J, Wang J, Xia K, Liang C, Fang W, Zhou C, Tao H. Escin induces caspase-dependent apoptosis and autophagy through the ROS/p38 MAPK signalling pathway in human osteosarcoma cells in vitro and in vivo. Cell Death Dis 2017; 8:e3113. [PMID: 29022891 PMCID: PMC5682655 DOI: 10.1038/cddis.2017.488] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/05/2017] [Accepted: 08/09/2017] [Indexed: 02/06/2023]
Abstract
Osteosarcoma is one of the most malignant neoplasms in adolescents, and it generally develops multidrug resistance. Escin, a natural mixture of triterpene saponins isolated from Aesculus hippocastanum (horse chestnut), has demonstrated potent anti-tumour potential in vitro and in vivo. In the present study, we found that escin inhibited osteosarcoma proliferation in a dose- and time-dependent manner. Additionally, escin-induced apoptosis was evidenced by the increased expression of caspase-related proteins and the formation of apoptotic bodies. Escin also induced autophagy, with elevated LC3, ATG5, ATG12 and Beclin expression as well as autophagosome formation. Inhibition of escin-induced autophagy promoted apoptosis. Moreover, p38 mitogen-activated protein kinases (MAPKs) and reactive oxygen species (ROS) were activated by escin. A p38 MAPK inhibitor partially attenuated the autophagy and apoptosis triggered by escin, but a ROS scavenger showed a greater inhibitory effect. Finally, the therapeutic efficacy of escin against osteosarcoma was demonstrated in an orthotopic model. Overall, escin counteracted osteosarcoma by inducing autophagy and apoptosis via the activation of the ROS/p38 MAPK signalling pathway; these findings provide evidence for escin as a novel and potent therapeutic for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Jian Zhu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Wei Yu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Bing Liu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Yitian Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Jianlin Shao
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Junjie Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Kaishun Xia
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Chengzhen Liang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Weijing Fang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Chenhe Zhou
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Huimin Tao
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| |
Collapse
|
42
|
Koppe T, Patchen B, Cheng A, Bhasin M, Vulpe C, Schwartz RE, Moreno‐Navarrete JM, Fernandez‐Real JM, Pissios P, Fraenkel PG. Nicotinamide N-methyltransferase expression decreases in iron overload, exacerbating toxicity in mouse hepatocytes. Hepatol Commun 2017; 1:803-815. [PMID: 29404495 PMCID: PMC5678920 DOI: 10.1002/hep4.1083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 07/27/2017] [Accepted: 08/01/2017] [Indexed: 12/19/2022] Open
Abstract
Iron overload causes the generation of reactive oxygen species that can lead to lasting damage to the liver and other organs. The goal of this study was to identify genes that modify the toxicity of iron overload. We studied the effect of iron overload on the hepatic transcriptional and metabolomic profile in mouse models using a dietary model of iron overload and a genetic model, the hemojuvelin knockout mouse. We then evaluated the correlation of nicotinamide N-methyltransferase (NNMT) expression with body iron stores in human patients and the effect of NNMT knockdown on gene expression and viability in primary mouse hepatocytes. We found that iron overload induced significant changes in the expression of genes and metabolites involved in glucose and nicotinamide metabolism and that NNMT, an enzyme that methylates nicotinamide and regulates hepatic glucose and cholesterol metabolism, is one of the most strongly down-regulated genes in the liver in both genetic and dietary iron overload. We found that hepatic NNMT expression is inversely correlated with serum ferritin levels and serum transferrin saturation in patients who are obese, suggesting that body iron stores regulate human liver NNMT expression. Furthermore, we demonstrated that adenoviral knockdown of NNMT in primary mouse hepatocytes exacerbates iron-induced hepatocyte toxicity and increases expression of transcriptional markers of oxidative and endoplasmic reticulum stress, while overexpression of NNMT partially reversed these effects. Conclusion: Iron overload alters glucose and nicotinamide transcriptional and metabolic pathways in mouse hepatocytes and decreases NNMT expression, while NNMT deficiency worsens the toxic effect of iron overload. For these reasons, NNMT may be a drug target for the prevention of iron-induced hepatotoxicity. (Hepatology Communications 2017;1:803-815).
Collapse
Affiliation(s)
- Tiago Koppe
- Division of Hematology/Oncology
- Cancer Research Institute, Beth Israel Deaconess Medical CenterBostonMA
- Department of MedicineHarvard Medical SchoolBostonMA
| | - Bonnie Patchen
- Division of Hematology/Oncology
- Cancer Research Institute, Beth Israel Deaconess Medical CenterBostonMA
- Department of MedicineHarvard Medical SchoolBostonMA
| | - Aaron Cheng
- Division of Hematology/Oncology
- Cancer Research Institute, Beth Israel Deaconess Medical CenterBostonMA
- Department of MedicineHarvard Medical SchoolBostonMA
| | - Manoj Bhasin
- Department of MedicineHarvard Medical SchoolBostonMA
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical CenterBostonMA
| | - Chris Vulpe
- Department of Physiological SciencesUniversity of FloridaGainesvilleFL
| | - Robert E. Schwartz
- Division of Gastroenterology and Hepatology, Weill Cornell Medical SchoolNew YorkNY
| | - Jose Maria Moreno‐Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de GironaHospital de Girona Dr. Josep Trueta and Universitat de GironaGironaSpain
- CIBER Fisopatologia de la Obesidad y Nutricion, Instituto de Salud Carlos IIIMadridSpain
| | - Jose Manuel Fernandez‐Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de GironaHospital de Girona Dr. Josep Trueta and Universitat de GironaGironaSpain
- CIBER Fisopatologia de la Obesidad y Nutricion, Instituto de Salud Carlos IIIMadridSpain
| | - Pavlos Pissios
- Division of Endocrinology, Beth Israel Deaconess Medical CenterBostonMA
| | - Paula G. Fraenkel
- Division of Hematology/Oncology
- Cancer Research Institute, Beth Israel Deaconess Medical CenterBostonMA
- Department of MedicineHarvard Medical SchoolBostonMA
| |
Collapse
|