1
|
Li J, Liu J, Tang Y, Zhang H, Zhang Y, Zha X, Zhao X. Role of C/EBP Homologous Protein (CHOP) and Nupr1 Interaction in Endoplasmic Reticulum Stress-Induced Apoptosis of Lens Epithelial Cells. Mol Biotechnol 2025; 67:1628-1640. [PMID: 38771421 PMCID: PMC11928426 DOI: 10.1007/s12033-024-01148-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/05/2024] [Indexed: 05/22/2024]
Abstract
Our study mainly analyzed the mechanism of C/EBP homologous protein (CHOP) and its interacting protein Nupr1 on endoplasmic reticulum stress (ERS) induced lens epithelial cells (LEC) apoptosis. Cell proliferation was detected by CCK-8. Apoptosis was detected by flow cytometry and TUNEL. Nupr1 expression was detected by RT-qPCR. The expressions of CHOP, Nupr1, apoptosis-related protein, and ERS-related protein were detected by Western blot. DCFH-DA probe was used to detect cell ROS. The SOD, GSH-PX, and MDA contents were detected by the kit. Co-IP was used to detect the interaction between CHOP and Nupr1. The morphology of the lens was detected by HE staining. The result shows that Tunicamycin (TU) can induce endoplasmic reticulum stress and apoptosis in LEC in a concentration-dependent manner. TU induction leads to the occurrence of CHOP nuclear translocation. Overexpression of CHOP can further enhance the inhibitory effect of TU on LEC proliferation and the promotion of apoptosis, while knockdown of CHOP has the opposite effect. CHOP and Nupr1 are interacting proteins, and knockdown of Nupr1 or addition of Nupr1 inhibitor ZZW-115 can reverse the effects of TU and overexpression of CHOP, respectively. It has been observed in animal experiments that treatment with oe-CHOP can further aggravate the pathological lesions of the rat lens, while ZZW-115 can reverse the effect of oe-CHOP to a certain extent and improve the lesions of the rat lens. Overall, CHOP interacts with Nupr1 to regulate apoptosis caused by ERS and mediate cataract progression in rats, and this study provides a new potential therapeutic target for the treatment of cataract.
Collapse
Affiliation(s)
- Jinghua Li
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Junyi Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Yongying Tang
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Hong Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Yuanping Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Xu Zha
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China.
| | - Xueying Zhao
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China.
| |
Collapse
|
2
|
Tang Y, Wang X, Huang M, Li Y, Liu X, Zeng H, Yang Y, Zhou M. Sports training improves motor function after spinal cord injury by regulating microtubule dynamics. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167587. [PMID: 39586504 DOI: 10.1016/j.bbadis.2024.167587] [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: 06/23/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Spinal cord injury (SCI) often results in persistent disabilities, primarily due to deficient axon regeneration and irreversible neuronal loss. Sports training is a widely adopted intervention in clinical practice and research to promote axonal sprouting and synaptic plasticity, thereby improving motor function after SCI. However, the precise mechanisms by which sports training improves motor function after SCI remain incompletely understood. We established a rat model of T9 spinal cord contusion and initiated sports training 1 week after SCI, which continued for eight weeks. Using transcriptome sequencing validated through western blotting and immunostaining, we demonstrated that sports training effectively reduced neuroinflammation and prevented neuronal loss. Furthermore, we discovered that sports training changed neuronal microtubule dynamics, facilitating axon regeneration and synaptic plasticity and ultimately improving motor function. These findings indicate that the modulation of neuronal microtubule dynamics may represent a critical mechanism through which sports training improves motor function after SCI.
Collapse
Affiliation(s)
- Yue Tang
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing 100191, China
| | - Xiaohuan Wang
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing 100191, China
| | - Mengjie Huang
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing 100191, China
| | - Yijie Li
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing 100191, China
| | - Xiaoxie Liu
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing 100191, China
| | - Hong Zeng
- Department of Rehabilitation Medicine, The Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Yanyan Yang
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing 100191, China.
| | - Mouwang Zhou
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing 100191, China.
| |
Collapse
|
3
|
Qiu H, Zhang M, Chen C, Wang H, Yue X. Decreasing β-Catenin Leads to Altered Endothelial Morphology, Increased Barrier Permeability and Cognitive Impairment During Chronic Methamphetamine Exposure. Int J Mol Sci 2025; 26:1514. [PMID: 40003980 PMCID: PMC11854931 DOI: 10.3390/ijms26041514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Cognitive impairment induced by chronic methamphetamine (METH) exposure exhibits similarities to neurodegenerative disorders and is associated with blood-brain barrier (BBB) dysfunction. However, the potential involvement of β-catenin in maintaining BBB integrity during METH exposure remains unexplored. In this study, Y-maze and novel object recognition tests were conducted to assess cognitive impairment in mice exposed chronically to methamphetamine for 2 and 4 weeks. Gd-DTPA and Evans blue leakage tests revealed disruption of the BBB in the hippocampus, while chronic METH exposure for 2 and 4 weeks significantly decreased β-catenin levels along with its transcriptionally regulated protein, claudin5. Additionally, various neural injury-related proteins, such as APP, Aβ1-42, p-tau (Thr181) and p-tau (Ser396), as well as neuroinflammation-related proteins, such as IL-6, IL-1β, and TNF-α, exhibited increased levels following chronic METH exposure. Furthermore, plasma analysis indicated elevated levels of p-Tau (total), neurofilament light chain, and GFAP. In vitro experiments demonstrated that exposure to METH resulted in dose-dependent and time-dependent reductions in cellular activity and connectivity of bEnd.3 and hcmec/D3 cells. Furthermore, β-catenin exhibited decreased levels and altered subcellular localization, transitioning from the cell membrane to the cytoplasm and nucleus upon METH exposure. Overexpression of β-catenin was found to alleviate endothelial toxicity and attenuate junctional weakening induced by METH. The aforementioned findings underscore the crucial involvement of β-catenin in endothelial cells during chronic METH exposure-induced disruption of the BBB, thereby presenting a potential novel target for addressing METH-associated cerebrovascular dysfunction and cognitive impairment.
Collapse
Affiliation(s)
| | | | | | - Huijun Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China; (H.Q.); (M.Z.); (C.C.)
| | - Xia Yue
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China; (H.Q.); (M.Z.); (C.C.)
| |
Collapse
|
4
|
Zhang M, Xu J, Liu Q, Yan X, Li N. TROP2 regulates cisplatin sensitivity of triple-negative breast cancer cells by regulating endoplasmic reticulum stress. Histol Histopathol 2025; 40:259-268. [PMID: 38884164 DOI: 10.14670/hh-18-771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Triple-negative breast cancer (TNBC) is a kind of breast cancer with a high metastasis rate and poor prognosis. As a transmembrane glycoprotein, tumor-associated calcium signal transducer 2 (TROP2) plays a certain role in the cancers. This study aimed to explore the potential mechanism of TROP2 affecting cisplatin (CDDP) resistance in TNBC from endoplasmic reticulum stress (ERS). MDA-MB-231 and CDDP-resistant cell lines MDA-MB-231/CDDP were used in this study, and the expression of TROP2 was detected by western blotting. After transfecting with the interference sequence of siRNA targeting TROP2, cell proliferation and apoptosis were detected by the cell counting kit-8, colony formation, and flow cytometry, and the expression of ERS-marker proteins was detected by western blotting. Furthermore, the effects of ERS in TROP2 on drug resistance of TNBC cells were explored by using ERS inhibitor 4-phenylbutyric acid (4-PBA). Results found that TROP2 expression in MDA-MB-231/CDDP was significantly upregulated compared with MDA-MB-231. The expression of TROP2 in MDA-MB-231/CDDP was significantly decreased after transfection with siRNA-TROP2, and the proliferation of MDA-MB-231 and MDA-MB-231/CDDP cells was significantly decreased after further induction with CDDP. TROP2 significantly affected TNBC cell cloning, apoptosis, and the expression of ERS-related marker proteins, while 4-PBA reversed the promoting effects of siRNA-TROP2 on apoptosis and ERS, as well as the inhibitory effects on cell proliferation, suggesting that TROP2 affected the resistance of TNBC cells to CDDP through ERS. In conclusion, TROP2 inhibited apoptosis of TNBC cells, improved the cell cloning ability, and regulated the sensitivity of TNBC cells to CDDP through ERS.
Collapse
Affiliation(s)
- Mingqi Zhang
- Department of Breast Surgery, Changzhi People's Hospital, Changzhi, China
| | - Jianzhong Xu
- Department of Breast Surgery, Changzhi People's Hospital, Changzhi, China
| | - Qing Liu
- Department of Emergency, Changzhi People's Hospital, Changzhi, China
| | - Xi Yan
- Department of Pharmacy, Changzhi People's Hospital, Changzhi, China
| | - Ning Li
- Department of Breast Surgery, Changzhi People's Hospital, Changzhi, China.
| |
Collapse
|
5
|
Ramli FF, Rejeki PS, Ibrahim N'I, Abdullayeva G, Halim S. A Mechanistic Review on Toxicity Effects of Methamphetamine. Int J Med Sci 2025; 22:482-507. [PMID: 39898237 PMCID: PMC11783064 DOI: 10.7150/ijms.99159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 11/26/2024] [Indexed: 02/04/2025] Open
Abstract
Persistent methamphetamine use causes many toxic effects in various organs, including the brain, heart, liver, kidney and eyes. The extent of its toxicity depends on numerous pharmacological factors, including route of administration, dose, genetic polymorphism related to drug metabolism and polysubstance abuse. Several molecular pathways have been proposed to activate oxidative stress, inflammation and apoptosis: B-cell lymphoma protein 2 (Bcl-2)-associated X (Bax)/Bcl2/caspase-3, nuclear factor erythroid 2-related factor (Nrf2)/heme oxygenase-1 (HO-1), protein kinase B (Akt)/mammalian target of rapamycin (mTOR)/p70S6K, trace amine-associated receptor 1 (TAAR1)/cAMP/lysyl oxidase, Sigmar1/ cAMP response element-binding protein (CREB)/mitochondrial fission-1 protein (Fis1), NADPH-Oxidase-2 (NOX-2), renal autophagy pathway, vascular endothelial growth factor (VEGF)/phosphatidylinositol-3-kinase (PI3K)/ protein kinase B (Akt)/endothelial nitric oxide synthase (eNOS), Nupr1/Chop/P53/PUMA/Beclin1 and Toll-like receptor (TLR)4/MyD88/TRAF6 pathways. The activation promotes pathological changes, including the disruption of the blood-brain barrier, myocardial infarction, cardiomyopathy, acute liver failure, acute kidney injury, chronic kidney disease, keratitis, retinopathy and vision loss. This review revisits the pharmacological profiles of methamphetamine and its effects on the brain, heart, liver, eyes, kidneys and endothelium. Understanding the mechanisms of methamphetamine toxicity is essential in developing treatment strategies to reverse or attenuate the progress of methamphetamine-associated organ damage.
Collapse
Affiliation(s)
- Fitri Fareez Ramli
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Kuala Lumpur, Malaysia
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Purwo Sri Rejeki
- Physiology Division, Department of Medical Physiology and Biochemistry, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Nurul 'Izzah Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Kuala Lumpur, Malaysia
| | - Gulnar Abdullayeva
- Old Road Campus Research Building, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
- Institute of Molecular Biology & Biotechnologies, Ministry of Science and Education of the Republic of Azerbaijan, 11 Izzat Nabiyev Str., AZ1073, Baku, Azerbaijan
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, OX3 7TY, UK
| | - Shariff Halim
- Faculty of Health Sciences, Universiti Teknologi MARA Cawangan Pulau Pinang, Kampus Bertam, 13200 Pulau Pinang, Malaysia
| |
Collapse
|
6
|
Latifi Z, Nikanfar S, Khodavirdilou R, Beirami SM, Khodavirdilou L, Fattahi A, Oghbaei F. MicroRNAs as diagnostic biomarkers in diabetes male infertility: a systematic review. Mol Biol Rep 2024; 52:90. [PMID: 39739064 DOI: 10.1007/s11033-024-10197-1] [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: 07/11/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025]
Abstract
This study conducts an in-depth review of the correlation between testis tissue changes and circulating microRNAs (miRNA) in diabetes-induced male reproductive complications, drawing upon both animal and clinical studies. The original articles published in English that specifically investigate miRNAs linked to male infertility in humans or animals with either type I or ΙΙ diabetes mellitus were included. The relevant articles were gathered from the PubMed, Google Scholar, Cochrane Library, and ScienceDirect databases. The quality of study was assessed utilizing the Joanna Briggs Institute (JBI) Critical Appraisal Checklist for Prevalence Studies. We collected an overall number of 1989 citations relating to our research subject. Following the elimination of articles based on the criteria, a total of 20 papers were included in the study. Aberrant expression profiles of 25 miRNAs were identified in diabetes associated with male reproductive issues, with 15 miRNAs exhibiting increased expression and 10 miRNAs showing decreased expression. Among the chosen publications, eighteen were identified as low-risk and two were classed as moderate quality. The dysregulated miRNAs were linked to testicular injury, disrupted steroid production, decreased sperm development and quality, and erectile dysfunction. The results demonstrate that the miRNA-mRNA network is linked to the pathological progression of diabetic testicular damage or erectile dysfunction. From a therapeutic perspective, the identification of circulating miRNAs could be beneficial in the timely identification and prevention of diabetes problems, such as diabetes-induced male infertility. Among all signaling pathways influenced by modified miRNAs, the Bax-caspase-3, MAPK, PI3K-Akt, and eNOS-cGMP-PKC were the main deregulated pathways.
Collapse
Affiliation(s)
- Zeinab Latifi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Saba Nikanfar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Rasa Khodavirdilou
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Sohrab Minaei Beirami
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Lida Khodavirdilou
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Amir Fattahi
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Farnaz Oghbaei
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
7
|
Tian Q, Zhou J, Xu Z, Wang B, Liao J, Duan K, Li X, Huang E, Xie WB. STIM1 Mediates Methamphetamine-Induced Neuronal Autophagy and Apoptosis. Neurotoxicology 2024; 103:S0161-813X(24)00061-5. [PMID: 38901802 DOI: 10.1016/j.neuro.2024.06.006] [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: 12/11/2023] [Revised: 03/18/2024] [Accepted: 06/10/2024] [Indexed: 06/22/2024]
Abstract
Methamphetamine (METH) is a widely abused amphetamine-type psychoactive drug that causes serious health problems. Previous studies have demonstrated that METH can induce neuron autophagy and apoptosis in vivo and in vitro. However, the molecular mechanisms underlying METH-induced neuron autophagy and apoptosis remain poorly understood. Stromal interacting molecule 1 (STIM1) was hypothesized to be involved in METH-induced neuron autophagy and apoptosis. Therefore, the expression of STIM1 protein was measured and the effect of blocking STIM1 expression with siRNA was investigated in cultured neuronal cells, and the hippocampus and striatum of mice exposed to METH. Furthermore, intracellular calcium concentration and endoplasmic reticulum (ER) stress-related proteins were determined in vitro and in vivo in cells treated with METH. The results suggested that STIM1 mediates METH-induced neuron autophagy by activating the p-Akt/p-mTOR pathway. METH exposure also resulted in increased expression of Orai1, which was reversed after STIM1 silencing. Moreover, the disruption of intracellular calcium homeostasis induced ER stress and up-regulated the expression of pro-apoptotic protein CCAAT/enhancer-binding protein homologous protein (CHOP), resulting in classic mitochondria apoptosis. METH exposure can cause neuronal autophagy and apoptosis by increasing the expression of STIM1 protein; thus, STIM1 may be a potential gene target for therapeutics in METH-caused neurotoxicity.
Collapse
Affiliation(s)
- Qin Tian
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Jie Zhou
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Zhenzhen Xu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Bin Wang
- Forensic Science Institute of Ganzhou Public Security Bureau, Ganzhou 341000, PR China
| | - Jiashun Liao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Ke Duan
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Xiaoting Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Enping Huang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Wei-Bing Xie
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
8
|
Zhou J, Guo D, Xu ZZ, Liao JS, Li XT, Duan K, Chen SY, Xie WB. Nupr1-mediated vascular smooth muscle cell phenotype transformation involved in methamphetamine induces pulmonary hypertension. Cell Biol Toxicol 2024; 40:13. [PMID: 38347241 PMCID: PMC10861617 DOI: 10.1007/s10565-024-09849-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/26/2024] [Indexed: 02/15/2024]
Abstract
AIMS Nuclear protein 1 (Nupr1) is a multifunctional stress-induced protein involved in the regulation of tumorigenesis, apoptosis, and autophagy. However, its role in pulmonary hypertension (PH) after METH exposure remains unexplored. In this study, we aimed to investigate whether METH can induce PH and describe the role and mechanism of Nupr1 in the development of PH. METHODS AND RESULTS Mice were made to induce pulmonary hypertension (PH) upon chronic intermittent treatment with METH. Their right ventricular systolic pressure (RVSP) was measured to assess pulmonary artery pressure. Pulmonary artery morphometry was determined by H&E staining and Masson staining. Nupr1 expression and function were detected in human lungs, mice lungs exposed to METH, and cultured pulmonary arterial smooth muscle cells (PASMCs) with METH treatment. Our results showed that chronic intermittent METH treatment successfully induced PH in mice. Nupr1 expression was increased in the cultured PASMCs, pulmonary arterial media from METH-exposed mice, and METH-ingested human specimens compared with control. Elevated Nupr1 expression promoted PASMC phenotype change from contractile to synthetic, which triggered pulmonary artery remodeling and resulted in PH formation. Mechanistically, Nupr1 mediated the opening of store-operated calcium entry (SOCE) by activating the expression of STIM1, thereby promoting Ca2+ influx and inducing phenotypic conversion of PASMCs. CONCLUSIONS Nupr1 activation could promote Ca2+ influx through STIM1-mediated SOCE opening, which promoted METH-induced pulmonary artery remodeling and led to PH formation. These results suggested that Nupr1 played an important role in METH-induced PH and might be a potential target for METH-related PH therapy.
Collapse
Affiliation(s)
- Jie Zhou
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Dan Guo
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Zhen-Zhen Xu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jia-Shun Liao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xiao-Ting Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Ke Duan
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Shi-You Chen
- Department of Surgery, Medical Pharmacology & Physiology, University of Missouri, Columbia, MO, 65212, USA
| | - Wei-Bing Xie
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
9
|
Vincent B, Shukla M. The Common Denominators of Parkinson's Disease Pathogenesis and Methamphetamine Abuse. Curr Neuropharmacol 2024; 22:2113-2156. [PMID: 37691228 PMCID: PMC11337683 DOI: 10.2174/1570159x21666230907151226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 09/12/2023] Open
Abstract
The pervasiveness and mortality associated with methamphetamine abuse have doubled during the past decade, suggesting a possible worldwide substance use crisis. Epitomizing the pathophysiology and toxicology of methamphetamine abuse proclaims severe signs and symptoms of neurotoxic and neurobehavioral manifestations in both humans and animals. Most importantly, chronic use of this drug enhances the probability of developing neurodegenerative diseases manifolds. Parkinson's disease is one such neurological disorder, which significantly and evidently not only shares a number of toxic pathogenic mechanisms induced by methamphetamine exposure but is also interlinked both structurally and genetically. Methamphetamine-induced neurodegeneration involves altered dopamine homeostasis that promotes the aggregation of α-synuclein protofibrils in the dopaminergic neurons and drives these neurons to make them more vulnerable to degeneration, as recognized in Parkinson's disease. Moreover, the pathologic mechanisms such as mitochondrial dysfunction, oxidative stress, neuroinflammation and decreased neurogenesis detected in methamphetamine abusers dramatically resemble to what is observed in Parkinson's disease cases. Therefore, the present review comprehensively cumulates a holistic illustration of various genetic and molecular mechanisms putting across the notion of how methamphetamine administration and intoxication might lead to Parkinson's disease-like pathology and Parkinsonism.
Collapse
Affiliation(s)
- Bruno Vincent
- Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Mayuri Shukla
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 10210, Bangkok, Thailand
| |
Collapse
|
10
|
Salimi H, Haghighi AH, Ababzadeh S, Marefati H, Abbasian S, Pond AL, Gentil P. Aerobic training and vitamin E administration ameliorates cardiac apoptosis markers in rats exposed to methamphetamine. Eur J Transl Myol 2023; 33:12112. [PMID: 38112583 PMCID: PMC10811645 DOI: 10.4081/ejtm.2023.12112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/21/2023] Open
Abstract
Methamphetamine (MA) abuse is related to risks to the cardiovascular system. The present study aimed to compare the effects of moderate-intensity aerobic training (MIAT) and vitamin E (Vit.E) supplementation on markers of cardiac apoptosis following MA exposure. Fifty-four rats were randomly divided into six groups. CON group did not receive MA, while the others received MA alone or in combination with MIAT, Vit. E, MIAT+Vit E, or paraffin (PAR). These groups received MA incrementally for 23 consecutive days. Vit.E and MIAT+Vit.E groups received vitamin E three times a week for six weeks. MIAT and MIAT+Vit.E groups exercised for 25-40 min. Immunohistochemical and gene expression analyses were performed on the heart tissues. Bax and TGF-β expression was significantly higher, while Bcl-2 and VEGF expression was significantly lower in the MA and PAR groups than in the other groups (p < 0.05). Bcl-2 and VEGF expression was higher, and Bax and TGF-β expression was significantly lower in the MIAT and MIAT+Vit.E groups than in the other groups (p < 0.05). In Vit.E treated groups, Bax and TGF-β expression were lower, and VEGF was higher than that in the MA and PAR groups, but higher than those in the CON, MIAT and MIAT+Vit.E groups. MA increased the expression of Bax and TGF-β, and decreased the expression of Bcl-2 and VEGF, suggesting increased cardiac apoptosis. In contrast, MIAT and Vit.E decreased the expression of Bax and TGF-β, suggesting a reduction in cardiac apoptosis induced by MA.
Collapse
Affiliation(s)
- Hamidreza Salimi
- Department of Exercise Physiology, Faculty of Sports Sciences, Hakim Sabzevari University, Sabzevar.
| | - Amir Hossein Haghighi
- Department of Exercise Physiology, Faculty of Sport Sciences, Hakim Sabzevari University, Sabzevar.
| | - Shima Ababzadeh
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran; Department of Tissue Engineering and Regenerative Medicine, Faculty of Medical Sciences, Qom University of Medical Sciences, Qom.
| | - Hamid Marefati
- Department of Exercise Physiology, Faculty of Sports Sciences, Hakim Sabzevari University, Sabzevar.
| | - Sadegh Abbasian
- Department of Sport Sciences, Khavaran Institute of Higher Education, Mashhad.
| | - Amber L Pond
- Anatomy, Southern Illinois University School of Medicine, Carbondale, IL.
| | - Paulo Gentil
- Hypertension League, Federal University of Goias, Brazil; College of Physical Education and Dance, Federal University of Goias.
| |
Collapse
|
11
|
Xu L, Li L, Chen Q, Huang Y, Chen X, Qiao D. The Role of Non-coding RNAs in Methamphetamine-Induced Neurotoxicity. Cell Mol Neurobiol 2023; 43:2415-2436. [PMID: 36752885 PMCID: PMC11410138 DOI: 10.1007/s10571-023-01323-x] [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: 11/30/2022] [Accepted: 01/20/2023] [Indexed: 02/09/2023]
Abstract
Methamphetamine (METH) is an amphetamine-type stimulant that is highly toxic to the central nervous system (CNS). Repeated intake of METH can lead to addiction, which has become a globalized problem, resulting in multiple public health and safety problems. Recently, the non-coding RNA (ncRNA) has been certified to play an essential role in METH addiction through various mechanisms. Herein, we mainly focused on three kinds of ncRNAs including long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), which are involved in neurotoxicity effects such as cognitive impairment, behavioral abnormalities, and psychiatric disorders due to METH abuse. In addition, differential expression (DE) ncRNAs also suggest that specific responses and sensitivity to METH neurotoxicity exist in different brain regions and cells. We summarized the relationships between the ncRNAs and METH-induced neurotoxicity and psychiatric disturbances, respectively, hoping to provide new perspectives and strategies for the prevention and treatment of METH abuse. Schematic diagram of the non-coding RNAs (ncRNAs) was involved in methamphetamine (METH)-induced neurotoxicity. The ncRNAs were involved in METH-induced blood-brain barrier disruption, neuronal, astrocyte, and microglial damage, and synaptic neurotransmission impairment. The study of ncRNAs is a hot spot in the future to further understand the neurotoxicity of METH and provide more favorable scientific support for clinical diagnosis and innovation of related treatments.
Collapse
Affiliation(s)
- Luyao Xu
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, South Shaitai Road #1023. 510515, Guangzhou, China
| | - Lingyue Li
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, South Shaitai Road #1023. 510515, Guangzhou, China
| | - Qianling Chen
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, South Shaitai Road #1023. 510515, Guangzhou, China
| | - Yuebing Huang
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, South Shaitai Road #1023. 510515, Guangzhou, China
| | - Xuebing Chen
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, South Shaitai Road #1023. 510515, Guangzhou, China.
| | - Dongfang Qiao
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, South Shaitai Road #1023. 510515, Guangzhou, China.
| |
Collapse
|
12
|
Liu S, Costa M. The role of NUPR1 in response to stress and cancer development. Toxicol Appl Pharmacol 2022; 454:116244. [PMID: 36116561 DOI: 10.1016/j.taap.2022.116244] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/26/2022] [Accepted: 09/09/2022] [Indexed: 10/31/2022]
Abstract
Stress contributes to the development of many human diseases, including cancer. Based on the source of stress, it can be divided into external stress, such as environmental carcinogens, chemicals, and radiation, and internal stress, like endoplasmic reticulum (ER) stress, hypoxia, and oxidative stress. Nuclear Protein 1 (NUPR1, p8 or Com-1) is a small, highly basic transcriptional regulator that participates in regulating a variety of cellular processes including DNA repair, ER stress, oxidative stress response, cell cycle, autophagy, apoptosis, ferroptosis and chromatin remodeling. A large number of studies have reported that NUPR1 expression can be stimulated rapidly in response to various stresses. Thus, NUPR1 is also known as a stress-response gene. Since the role of NUPR1 in breast cancer was identified in 1999, an increasing number of studies sought to reveal its function in cancer. High expression of NUPR1 has been identified in oral squamous cell carcinoma, breast cancer, lung cancer, multiple myeloma, liver cancer and renal cancer. In this review, we summarize current studies of NUPR1 in response to multiple external stressors and internal stressors, and its role in mediating stressors to cause different cell signaling responses. In addition, this review discusses the function of NUPR1 in carcinogenesis, tumorigenesis, metastasis, and cancer therapy. Thus, this review gives a comprehensive insight into the role of NUPR1 in mediating signals from stress to different cell responses, and this process plays a role in the development of cancer.
Collapse
Affiliation(s)
- Shan Liu
- Division of Environmental Medicine, Dept of Medicine, New York University School of Medicine, NY, USA.
| | - Max Costa
- Division of Environmental Medicine, Dept of Medicine, New York University School of Medicine, NY, USA.
| |
Collapse
|
13
|
Luo BY, Zhou J, Guo D, Yang Q, Tian Q, Cai DP, Zhou RM, Xu ZZ, Wang HJ, Chen SY, Xie WB. Methamphetamine induces thoracic aortic aneurysm/dissection through C/EBPβ. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166447. [PMID: 35643386 PMCID: PMC9753351 DOI: 10.1016/j.bbadis.2022.166447] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/02/2022] [Accepted: 05/12/2022] [Indexed: 01/25/2023]
Abstract
AIMS Thoracic aortic aneurysm/dissection (TAAD) is a life-threatening disease with diverse clinical manifestations. Although the association between methamphetamine (METH) and TAAD is frequently observed, the causal relationship between METH abuse and aortic aneurysm/dissection has not been established. This study was designed to determine if METH causes aortic aneurysm/dissection and delineate the underlying mechanism. METHODS AND RESULTS A new TAAD model was developed by exposing METH to SD rats pre-treated with lysyl oxidase inhibitor β-aminopropionitrile (BAPN). Combination of METH and BAPN caused thoracic aortic aneurysm/dissection in 60% of rats. BAPN+METH significantly increased the expression and activities of both matrix metalloproteinase MMP2 and MMP9, consistent with the severe elastin breakage and dissection. Mechanistically, METH increased CCAAT-enhancer binding protein β (C/EBPβ) expression by enhancing mothers against decapentaplegic homolog 3 (Smad3) and extracellular regulated protein kinase (ERK1/2) signaling. METH also promoted C/EBPβ binding to MMP2 and MMP9 promoters. Blocking C/EBPβ significantly attenuated METH+BAPN-induced TAAD and MMP2/MMP9 expression. Moreover, BAPN+METH promoted aortic medial smooth muscle cell (SMC) apoptosis through C/EBPβ-mediated IGFBP5/p53/PUMA signaling pathways. More importantly, the expression of C/EBPβ, MMP2/MMP9, and apoptosis-promoting proteins was increased in the aorta of human patients with thoracic aortic dissection, suggesting that the mechanisms identified in animal study could be relevant to human disease. CONCLUSIONS Our study demonstrated that METH exposure has a casual effect on TAAD. C/EBPβ mediates METH-introduced TAAD formation by causing elastin breakage, medial cell loss and degeneration. Therefore, C/EBPβ may be a potential factor for TAAD clinical diagnosis or treatment.
Collapse
Affiliation(s)
- Bao-Ying Luo
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China; Zhangzhou Health Vocational College, Zhangzhou 363000, PR China
| | - Jie Zhou
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Dan Guo
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Qian Yang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Qin Tian
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Dun-Peng Cai
- Department of Surgery, Medical Pharmacology & Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Rui-Mei Zhou
- Department of Surgery, Medical Pharmacology & Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Zhen-Zhen Xu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Hui-Jun Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China.
| | - Shi-You Chen
- Department of Surgery, Medical Pharmacology & Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| | - Wei-Bing Xie
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China; NHC Key Laboratory of Drug Addiction Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China.
| |
Collapse
|
14
|
He S, Yao Y, Yang N, Wang Y, Liu D, Cao Z, Chen H, Fu Y, Yang M, Wang S, He G, Zhao Q. Dapagliflozin Protects Methamphetamine-Induced Cardiomyopathy by Alleviating Mitochondrial Damage and Reducing Cardiac Function Decline in a Mouse Model. Front Pharmacol 2022; 13:925276. [PMID: 35873593 PMCID: PMC9301370 DOI: 10.3389/fphar.2022.925276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Methamphetamine (METH)-induced cardiovascular toxicity has been attributed to its destructive effect on mitochondrial function at least to some extent. Previous studies highlighted the benefits of dapagliflozin (DAPA) on the cardiovascular system, but the response of METH-induced cardiomyopathy to DAPA is never addressed before. The present study aimed to investigate the potential ability of DAPA in preventing METH-induced cardiomyopathy.Materials and Methods: C57BL/6 mice were randomly divided into control group (n = 24), METH group (n = 24), and METH + DAPA group (n = 24). The METH-induced cardiomyopathy group received intraperitoneal METH injections at gradually increasing doses thrice weekly for 14 weeks. Mice in the METH + DAPA group were simultaneously treated with DAPA 1 mg/kg/day by intragastric administration. Echocardiography was performed to assess cardiac function. Reactive oxygen species (ROS), JC-1, and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assays were performed to evaluate oxidative stress, mitochondrial damage, and apoptosis, respectively. Mitochondrial and apoptosis-related protein expression was measured by western blotting.Results: Mice exposed to METH exhibited reduced cardiac function (left ventricular ejection fraction [LVEF]: 56.51 ± 6.49 vs. 73.62 ± 1.42, p < 0.01), fibrotic remodeling, and mitochondrial dysfunction, leading to apoptosis (apoptotic cells%: 7.4 ± 1.3 vs. 1.3 ± 0.5, p < 0.01). DAPA significantly reduced mitochondrial dynamics and function, ROS, apoptosis (apoptotic cells%: 2.4 ± 0.8 vs. 7.4 ± 1.3, p < 0.01), cardiac function decline (LVEF: 70.99 ± 4.936 vs. 56.51 ± 6.49, p < 0.01), and fibrotic remodeling. These results indicated that DAPA could be considered as an effective therapeutic agent in the protection against METH-associated cardiomyopathy.Conclusion: DAPA protects against METH-induced cardiomyopathy in mice by decreasing mitochondrial damage and apoptosis.
Collapse
Affiliation(s)
- Shanqing He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yajun Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Nan Yang
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Youcheng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Dishiwen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Huiyu Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yuntao Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Songjun Wang
- Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Guangjie He
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- *Correspondence: Qingyan Zhao,
| |
Collapse
|
15
|
Tsampoula M, Tarampoulous I, Manolakou T, Ninou E, Politis PK. The neurodevelopmental disorders associated gene Rnf113a regulates survival and differentiation properties of neural stem cells. Stem Cells 2022; 40:678-690. [DOI: 10.1093/stmcls/sxac030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 03/23/2022] [Indexed: 11/15/2022]
Abstract
Abstract
RNF113A (Ring Finger Protein 113A) is genetically associated with autism spectrum disorders and X-linked trichothiodystrophy (TTD) syndrome. Loss-of-function mutations in human RNF113A are causally linked to TTD, which is characterized by abnormal development of central nervous system (CNS) and mental retardation. How loss of RNF113A activity affects brain development is not known. Here we identify Rnf113a1 as a critical regulator of cell death and neurogenesis during mouse brain development. Rnf113a1 gene exhibits widespread expression in the embryonic CNS. Knockdown studies in embryonic cortical neural stem/progenitor cells (NSCs) and the mouse cortex suggest that Rnf113a1 controls survival, proliferation and differentiation properties of progenitor cells. Importantly, Rnf113a1 deficiency triggers cell apoptosis via a combined action on essential regulators of cell survival, including p53, Nupr1 and Rad51. Collectively, these observations establish Rnf113a1 as a regulatory factor in CNS development and provide insights for its role in neurodevelopmental defects associated with TTD and autism.
Collapse
Affiliation(s)
- Matina Tsampoula
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Isaak Tarampoulous
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Theodora Manolakou
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Elpinickie Ninou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| |
Collapse
|
16
|
Contreras L, Rodríguez-Gil A, Muntané J, de la Cruz J. Broad Transcriptomic Impact of Sorafenib and Its Relation to the Antitumoral Properties in Liver Cancer Cells. Cancers (Basel) 2022; 14:cancers14051204. [PMID: 35267509 PMCID: PMC8909169 DOI: 10.3390/cancers14051204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/21/2022] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is the fourth most frequent cause of cancer-related mortality worldwide. While ablation, resection and orthotopic liver transplantation are indicated at an early stage of the disease, Sorafenib (Sfb) is the current most administrated first-line treatment for advanced HCC, even though its therapeutic benefit is limited due to the appearance of resistance. Deep knowledge on the molecular consequences of Sfb-treatment is essentially required for optimizing novel therapeutic strategies to improve the outcomes for patients with advanced HCC. In this study, we analyzed differential gene expression changes in two well characterized liver cancer cell lines upon a Sfb-treatment, demonstrating that both lines responded similarly to the treatment. Our results provide valuable information on the molecular action of Sfb on diverse cellular fundamental processes such as DNA repair, translation and proteostasis and identify rationalization issues that could provide a different therapeutic perspective to Sfb. Abstract Hepatocellular carcinoma (HCC) is one of the most frequent and essentially incurable cancers in its advanced stages. The tyrosine kinase inhibitor Sorafenib (Sfb) remains the globally accepted treatment for advanced HCC. However, the extent of its therapeutic benefit is limited. Sfb exerts antitumor activity through its cytotoxic, anti-proliferative and pro-apoptotic roles in HCC cells. To better understand the molecular mechanisms underlying these effects, we used RNA sequencing to generate comprehensive transcriptome profiles of HepG2 and SNU423, hepatoblastoma- (HB) and HCC-derived cell lines, respectively, following a Sfb treatment at a pharmacological dose. This resulted in similar alterations of gene expression in both cell lines. Genes functionally related to membrane trafficking, stress-responsible and unfolded protein responses, circadian clock and activation of apoptosis were predominantly upregulated, while genes involved in cell growth and cycle, DNA replication and repair, ribosome biogenesis, translation initiation and proteostasis were downregulated. Our results suggest that Sfb causes primary effects on cellular stress that lead to upregulation of selective responses to compensate for its negative effect and restore homeostasis. No significant differences were found specifically affecting each cell line, indicating the robustness of the Sfb mechanism of action despite the heterogeneity of liver cancer. We discuss our results on terms of providing rationalization for possible strategies to improve Sfb clinical outcomes.
Collapse
Affiliation(s)
- Laura Contreras
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, E-41013 Seville, Spain; (L.C.); (A.R.-G.)
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, E-41012 Seville, Spain
| | - Alfonso Rodríguez-Gil
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, E-41013 Seville, Spain; (L.C.); (A.R.-G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), E-28029 Madrid, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, E-41009 Sevilla, Spain
| | - Jordi Muntané
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, E-41013 Seville, Spain; (L.C.); (A.R.-G.)
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, E-41009 Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), E-28029 Madrid, Spain
- Correspondence: (J.M.); (J.d.l.C.); Tel.: +34-955-923-122 (J.M.); +34-923-126 (J.d.l.C.)
| | - Jesús de la Cruz
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, E-41013 Seville, Spain; (L.C.); (A.R.-G.)
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, E-41012 Seville, Spain
- Correspondence: (J.M.); (J.d.l.C.); Tel.: +34-955-923-122 (J.M.); +34-923-126 (J.d.l.C.)
| |
Collapse
|
17
|
Dong G, Zhang R, Hu Q, Martin EM, Qin Y, Lu C, Xia Y, Wang X, Du G. Prothioconazole induces cell cycle arrest by up-regulation of EIF4EBP1 in extravillous trophoblast cells. Arch Toxicol 2022; 96:559-570. [PMID: 35048155 DOI: 10.1007/s00204-021-03203-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/09/2021] [Indexed: 11/30/2022]
Abstract
Prothioconazole (PTC) is a new broad-spectrum triazole antibacterial agent that is being widely used in agriculture. PTC has been linked to a number of reproductive outcomes including embryo implantation disorder; however, the exact mechanism underlying this relationship has yet to be determined. Proper trophoblast proliferation and migration is a prerequisite for successful embryo implantation. To elucidate the underlying molecular perturbations, we detect the effect of PTC on extravillous trophoblast cells proliferation and migration, and investigate its potential mechanisms. Exposure to different concentrations of PTC (0-500 μM) significantly inhibited the cell viability and migration ability (5 μM PTC exposure), and also caused the cell cycle arrest at the lowest dose (1 μM PTC exposure). Transcriptome analysis revealed that PTC exposure disturbed multiple biological processes including cell cycle and apoptosis, consistent with cell phenotype. Specifically, eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1, 4E-BP1) was identified as up-regulated in PTC exposure group and knockdown of EIF4EBP1, and attenuated the G1 phase arrest induced by PTC exposure. In summary, our data demonstrated that 4E-BP1 participated in PTC-induced cell cycle arrest in extravillous trophoblast cells by regulating cyclin D1. These findings shed light on the potential adverse effect of PTC exposure on the embryo implantation.
Collapse
Affiliation(s)
- Guangzhu Dong
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Road, Nanjing, 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Baijiahu Community Health Service Center, Moling Street, Jiangning District, Nanjing, 211102, China
| | - Rui Zhang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Road, Nanjing, 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Qi Hu
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Road, Nanjing, 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Center for Disease Control and Prevention, Beilun District, Ningbo, 315899, China
| | - Elizabeth M Martin
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, 27709, USA
- Department of Health and Human Services, Postdoctoral Research Associate Training Program, National Institute of General Medical Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Yufeng Qin
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Department of Microbes and Infection, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chuncheng Lu
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Road, Nanjing, 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Road, Nanjing, 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Road, Nanjing, 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Guizhen Du
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Road, Nanjing, 211166, China.
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
18
|
Xia Q, Zhan G, Mao M, Zhao Y, Li X. TRIM45 causes neuronal damage by aggravating microglia-mediated neuroinflammation upon cerebral ischemia and reperfusion injury. Exp Mol Med 2022; 54:180-193. [PMID: 35217833 PMCID: PMC8894463 DOI: 10.1038/s12276-022-00734-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/06/2021] [Accepted: 12/15/2021] [Indexed: 12/29/2022] Open
Abstract
Excessive and unresolved neuroinflammation is a key component of the pathological cascade in brain injuries such as ischemic stroke. Tripartite motif-containing 45 (TRIM45) is a ubiquitin E3 ligase involved in various critical biological processes. However, the role of TRIM45 in cerebral ischemia remains unknown. Here, we found that the TRIM45 protein was highly expressed in the peri-infarct areas of mice subjected to cerebral ischemia and reperfusion injury induced by middle cerebral artery occlusion. This study systemically evaluated the putative role of TRIM45 in the regulation of neuroinflammation during ischemic injury and the potential underlying mechanisms. We found that TRIM45 knockdown significantly decreased proinflammatory cytokine and chemokine production in primary cultured microglia challenged with oxygen-glucose deprivation and reoxygenation (OGD/R) treatment. Mechanistically, we demonstrated that TRIM45 constitutively interacted with TAB2 and consequently facilitated the Lys-63-linked polyubiquitination of TAB2, leading to the formation of the TAB1-TAK1-TAB2 complex and activation of TAK1, which was ultimately followed by activation of the nuclear factor-kappa B (NF-κB) signaling pathway. In an in vitro coculture Transwell system, downregulation of TRIM45 expression also inhibited the OGD/R-induced activation of microglia and alleviated neuronal apoptosis. More importantly, microglia-specific knockdown of TRIM45 in mice significantly reduced the infarct size, mitigated neurological deficit scores, and improved cognitive function after ischemic stroke. Taken together, our study reveals that the TRIM45-TAB2 axis is a crucial checkpoint that controls NF-κB signaling in microglia during cerebral ischemia and reperfusion injury. Therefore, targeting TRIM45 may be an attractive therapeutic strategy.
Collapse
Affiliation(s)
- Qian Xia
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Gaofeng Zhan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Meng Mao
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xing Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
19
|
Jung I, Park M, Jeong MH, Park K, Kim WH, Kim GY. Transcriptional analysis of gasoline engine exhaust particulate matter 2.5-exposed human umbilical vein endothelial cells reveals the different gene expression patterns related to the cardiovascular diseases. Biochem Biophys Rep 2022; 29:101190. [PMID: 34988296 PMCID: PMC8695280 DOI: 10.1016/j.bbrep.2021.101190] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 11/28/2022] Open
Abstract
Particulate matter (PM) causes several diseases, including cardiovascular diseases (CVDs). Previous studies compared the gene expression patterns in airway epithelial cells and keratinocytes exposed to PM. However, analysis of differentially expressed gene (DEGs) in endothelial cells exposed to PM2.5 (diameter less than 2.5 μm) from fossil fuel combustion has been limited. Here, we exposed human umbilical vein endothelial cells (HUVECs) to PM2.5 from combustion of gasoline, performed RNA-seq analysis, and identified DEGs. Exposure to the IC50 concentrations of gasoline engine exhaust PM2.5 (GPM) for 24 h yielded 1081 (up-regulation: 446, down-regulation: 635) DEGs. The most highly up-regulated gene is NGFR followed by ADM2 and NUPR1. The most highly down-regulated gene is TNFSF10 followed by GDF3 and EDN1. Gene Ontology enrichment analysis revealed that GPM regulated genes involved in cardiovascular system development, tube development and circulatory system development. Kyoto Encyclopedia of Genes and Genomes and Reactome pathway analyses showed that genes related to cytokine–cytokine receptor interactions and cytokine signaling in the immune system were significantly affected by GPM. We confirmed the RNA-seq data of some highly altered genes by qRT-PCR and showed the induction of NGFR, ADM2 and IL-11 at a protein level, indicating that the observed gene expression patterns were reliable. Given the adverse effects of PM2.5 on CVDs, our findings provide new insight into the importance of several DEGs and pathways in GPM-induced CVDs.
Collapse
Affiliation(s)
- Inkyo Jung
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Minhan Park
- School of Earth Science and Environmental Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Myong-Ho Jeong
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Kihong Park
- School of Earth Science and Environmental Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Won-Ho Kim
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Geun-Young Kim
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| |
Collapse
|
20
|
Multiparametric Evaluation of Post-MI Small Animal Models Using Metabolic ([ 18F]FDG) and Perfusion-Based (SYN1) Heart Viability Tracers. Int J Mol Sci 2021; 22:ijms222212591. [PMID: 34830471 PMCID: PMC8619497 DOI: 10.3390/ijms222212591] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 12/01/2022] Open
Abstract
Cardiovascular diseases (CVD), with myocardial infarction (MI) being one of the crucial components, wreak havoc in developed countries. Advanced imaging technologies are required to obtain quick and widely available diagnostic data. This paper describes a multimodal approach to in vivo perfusion imaging using the novel SYN1 tracer based on the fluorine-18 isotope. The NOD-SCID mice were injected intravenously with SYN1 or [18F] fluorodeoxyglucose ([18F]-FDG) radiotracers after induction of the MI. In all studies, the positron emission tomography–computed tomography (PET/CT) technique was used. To obtain hemodynamic data, mice were subjected to magnetic resonance imaging (MRI). Finally, the biodistribution of the SYN1 compound was performed using Wistar rat model. SYN1 showed normal accumulation in mouse and rat hearts, and MI hearts correctly indicated impaired cardiac segments when compared to [18F]-FDG uptake. In vivo PET/CT and MRI studies showed statistical convergence in terms of the size of the necrotic zone and cardiac function. This was further supported with RNAseq molecular analyses to correlate the candidate function genes’ expression, with Serpinb1c, Tnc and Nupr1, with Trem2 and Aldolase B functional correlations showing statistical significance in both SYN1 and [18F]-FDG. Our manuscript presents a new fluorine-18-based perfusion radiotracer for PET/CT imaging that may have importance in clinical applications. Future research should focus on confirmation of the data elucidated here to prepare SYN1 for first-in-human trials.
Collapse
|
21
|
Tobeiha M, Rajabi A, Raisi A, Mohajeri M, Yazdi SM, Davoodvandi A, Aslanbeigi F, Vaziri M, Hamblin MR, Mirzaei H. Potential of natural products in osteosarcoma treatment: Focus on molecular mechanisms. Biomed Pharmacother 2021; 144:112257. [PMID: 34688081 DOI: 10.1016/j.biopha.2021.112257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/21/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is the most frequent type of bone cancer found in children and adolescents, and commonly arises in the metaphyseal region of tubular long bones. Standard therapeutic approaches, such as surgery, chemotherapy, and radiation therapy, are used in the management of osteosarcoma. In recent years, the mortality rate of osteosarcoma has decreased due to advances in treatment methods. Today, the scientific community is investigating the use of different naturally derived active principles against various types of cancer. Natural bioactive compounds can function against cancer cells in two ways. Firstly they can act as classical cytotoxic compounds by non-specifically affecting macromolecules, such as DNA, enzymes, and microtubules, which are also expressed in normal proliferating cells, but to a greater extent by cancer cells. Secondly, they can act against oncogenic signal transduction pathways, many of which are activated in cancer cells. Some bioactive plant-derived agents are gaining increasing attention because of their anti-cancer properties. Moreover, some naturally-derived compounds can significantly promote the effectiveness of standard chemotherapy drugs, and in certain cases are able to ameliorate drug-induced adverse effects caused by chemotherapy. In the present review we summarize the effects of various naturally-occurring bioactive compounds against osteosarcoma.
Collapse
Affiliation(s)
- Mohammad Tobeiha
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Rajabi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Arash Raisi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahshad Mohajeri
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Amirhossein Davoodvandi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fatemeh Aslanbeigi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - MohamadSadegh Vaziri
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
22
|
Tan X, Cai D, Chen N, Du S, Qiao D, Yue X, Wang T, Li J, Xie W, Wang H. Methamphetamine mediates apoptosis of vascular smooth muscle cells via the chop-related endoplasmic reticulum stress pathway. Toxicol Lett 2021; 350:98-110. [PMID: 34214594 DOI: 10.1016/j.toxlet.2021.06.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/14/2022]
Abstract
Methamphetamine (METH) is a highly addictive amphetamine-type drug that has caused persistent harm to society and human health in recent years. Most studies have shown that METH severely damages the central nervous system, and this drug has been found to be toxic to the cardiovascular system in recent years. Therefore, we hypothesized that METH may also damage vascular smooth muscle. We examined the expression of the apoptosis-related proteins Caspase 3 and PARP after METH treatment in vivo and in vitro and detected the expression of endoplasmic reticulum stress-related proteins. After treatment with the endoplasmic reticulum stress inhibitor 4-PBA, changes in the above indicators were examined. C/EBP homologous protein (Chop) expression was also detected, and the relationship between endoplasmic reticulum stress and apoptosis was further determined by siRNA silencing of Chop. The results indicated that METH can induce apoptosis of vascular smooth muscle cells (VSMCs) and upregulate the expression of Chop and endoplasmic reticulum stress-related proteins. Chop inhibits protein kinase B phosphorylation and further inhibits forkhead box class O3a (Foxo3a) dephosphorylation, resulting in increased p53 upregulated molecular of apoptosis (PUMA) transcription. Increased PUMA induces apoptosis through the mitochondrial pathway. These results indicate that Chop is involved in the METH-induced endoplasmic reticulum stress and apoptosis in VSMCs and may be a potential therapeutic target for METH-induced VSMC injury.
Collapse
Affiliation(s)
- Xiaohui Tan
- School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Dunpeng Cai
- School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Na Chen
- Department of Pathology, Guangdong Women and Children Hospital, Guangzhou, 511400, Guangdong, China
| | - Sihao Du
- School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Dongfang Qiao
- School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xia Yue
- School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Tao Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jia Li
- School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Weibing Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China; Nanhai Hospital, Southern Medical University, Foshan, 528244, Guangdong, China.
| |
Collapse
|
23
|
Jayanthi S, Daiwile AP, Cadet JL. Neurotoxicity of methamphetamine: Main effects and mechanisms. Exp Neurol 2021; 344:113795. [PMID: 34186102 PMCID: PMC8338805 DOI: 10.1016/j.expneurol.2021.113795] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/03/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022]
Abstract
Methamphetamine (METH) is an illicit psychostimulant that is abused throughout the world. METH addiction is also a major public health concern and the abuse of large doses of the drug is often associated with serious neuropsychiatric consequences that may include agitation, anxiety, hallucinations, paranoia, and psychosis. Some human methamphetamine users can also suffer from attention, memory, and executive deficits. METH-associated neurological and psychiatric complications might be related, in part, to METH-induced neurotoxic effects. Those include altered dopaminergic and serotonergic functions, neuronal apoptosis, astrocytosis, and microgliosis. Here we have endeavored to discuss some of the main effects of the drug and have presented the evidence supporting certain of the molecular and cellular bases of METH neurotoxicity. The accumulated evidence suggests the involvement of transcription factors, activation of dealth pathways that emanate from mitochondria and endoplasmic reticulum (ER), and a role for neuroinflammatory mechanisms. Understanding the molecular processes involved in METH induced neurotoxicity should help in developing better therapeutic approaches that might also serve to attenuate or block the biological consequences of use of large doses of the drug by some humans who meet criteria for METH use disorder.
Collapse
Affiliation(s)
- Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, United States of America
| | - Atul P Daiwile
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, United States of America
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, United States of America.
| |
Collapse
|
24
|
Augello G, Emma MR, Azzolina A, Puleio R, Condorelli L, Cusimano A, Giannitrapani L, McCubrey JA, Iovanna JL, Cervello M. The NUPR1/p73 axis contributes to sorafenib resistance in hepatocellular carcinoma. Cancer Lett 2021; 519:250-262. [PMID: 34314755 DOI: 10.1016/j.canlet.2021.07.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/24/2021] [Accepted: 07/19/2021] [Indexed: 01/10/2023]
Abstract
The multikinase inhibitor sorafenib was the first drug approved by the FDA for treating patients with advanced hepatocellular carcinoma (HCC). However, sorafenib resistance remains a major challenge for improving the effectiveness of HCC treatment. Previously, we identified several genes modulated after sorafenib treatment of human HCC cells, including the stress-inducible nuclear protein 1 (NUPR1) gene. Multiple studies have shown that NUPR1 regulates autophagy, apoptosis, and chemoresistance. Here, we demonstrate that treatment of HCC cells with sorafenib resulted in the activation of autophagic flux. NUPR1 knock-down (KD) in HCC cells was associated with increased p62 expression, suggesting an impairment of autophagic flux, and with a significant increase of cell sensitivity to sorafenib. In NUPR1 KD cells, reduced levels of NUPR1 were associated with the increased expression of p73 as well as its downstream transcription targets PUMA, NOXA, and p21. Simultaneous silencing of p73 and NUPR1 in HCC cells resulted in increased resistance to sorafenib, as compared to the single KD of either gene. Conversely, pharmacological activation of p73, via the novel p73 small molecule activator NSC59984, determined synergistic anti-tumor effects in sorafenib-treated HCC cells. The combination of NSC59984 and sorafenib, when compared to either treatment alone, synergistically suppressed tumor growth of HCC cells in vivo. Our data suggest that the activation of the p73 pathway achieved by NUPR1 KD potentiates sorafenib-induced anti-tumor effects in HCC cells. Moreover, combined pharmacological therapy with the p73 activator NSC59984 and sorafenib could represent a novel approach for HCC treatment.
Collapse
Affiliation(s)
- Giuseppa Augello
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Maria Rita Emma
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Antonina Azzolina
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Roberto Puleio
- Istituto Zooprofilattico Sperimentale Della Sicilia "A. Mirri", Palermo, Italy
| | - Lucia Condorelli
- Istituto Zooprofilattico Sperimentale Della Sicilia "A. Mirri", Palermo, Italy
| | - Antonella Cusimano
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Lydia Giannitrapani
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy; Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA
| | - Juan Lucio Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Melchiorre Cervello
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy.
| |
Collapse
|
25
|
Huang C, Santofimia-Castaño P, Iovanna J. NUPR1: A Critical Regulator of the Antioxidant System. Cancers (Basel) 2021; 13:cancers13153670. [PMID: 34359572 PMCID: PMC8345110 DOI: 10.3390/cancers13153670] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Nuclear protein 1 (NUPR1) is activated in cellular stress and is expressed at high levels in cancer cells. Much evidence has been gathered supporting its critical role in regulating the antioxidant system. Our review aims to summarize the literature data on the impact of NUPR1 on the oxidative stress response via such a regulatory role and how its inhibition induces reactive oxygen species-mediated cell death, such as ferroptosis. Abstract Nuclear protein 1 (NUPR1) is a small intrinsically disordered protein (IDP) activated in response to various types of cellular stress, including endoplasmic reticulum (ER) stress and oxidative stress. Reactive oxygen species (ROS) are mainly produced during mitochondrial oxidative metabolism, and directly impact redox homeostasis and oxidative stress. Ferroptosis is a ROS-dependent programmed cell death driven by an iron-mediated redox reaction. Substantial evidence supports a maintenance role of the stress-inducible protein NUPR1 on cancer cell metabolism that confers chemotherapeutic resistance by upregulating mitochondrial function-associated genes and various antioxidant genes in cancer cells. NUPR1, identified as an antagonist of ferroptosis, plays an important role in redox reactions. This review summarizes the current knowledge on the mechanism behind the observed impact of NUPR1 on mitochondrial function, energy metabolism, iron metabolism, and the antioxidant system. The therapeutic potential of genetic or pharmacological inhibition of NUPR1 in cancer is also discussed. Understanding the role of NUPR1 in the antioxidant system and the mechanisms behind its regulation of ferroptosis may promote the development of more efficacious strategies for cancer therapy.
Collapse
|
26
|
Dynamin-related protein 1 deficiency accelerates lipopolysaccharide-induced acute liver injury and inflammation in mice. Commun Biol 2021; 4:894. [PMID: 34290349 PMCID: PMC8295278 DOI: 10.1038/s42003-021-02413-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 06/30/2021] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial fusion and fission, which are strongly related to normal mitochondrial function, are referred to as mitochondrial dynamics. Mitochondrial fusion defects in the liver cause a non-alcoholic steatohepatitis-like phenotype and liver cancer. However, whether mitochondrial fission defect directly impair liver function and stimulate liver disease progression, too, is unclear. Dynamin-related protein 1 (DRP1) is a key factor controlling mitochondrial fission. We hypothesized that DRP1 defects are a causal factor directly involved in liver disease development and stimulate liver disease progression. Drp1 defects directly promoted endoplasmic reticulum (ER) stress, hepatocyte death, and subsequently induced infiltration of inflammatory macrophages. Drp1 deletion increased the expression of numerous genes involved in the immune response and DNA damage in Drp1LiKO mouse primary hepatocytes. We administered lipopolysaccharide (LPS) to liver-specific Drp1-knockout (Drp1LiKO) mice and observed an increased inflammatory cytokine expression in the liver and serum caused by exaggerated ER stress and enhanced inflammasome activation. This study indicates that Drp1 defect-induced mitochondrial dynamics dysfunction directly regulates the fate and function of hepatocytes and enhances LPS-induced acute liver injury in vivo.
Collapse
|
27
|
Borrello MT, Santofimia-Castaño P, Bocchio M, Listi A, Fraunhoffer N, Soubeyran P, Chevet E, Pin C, Iovanna J. NUPR1 interacts with eIF2α and is required for resolution of the ER stress response in pancreatic tissue. FEBS J 2021; 288:4081-4097. [PMID: 33403797 DOI: 10.1111/febs.15700] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 11/20/2020] [Accepted: 01/04/2021] [Indexed: 12/22/2022]
Abstract
Nuclear protein 1 (NUPR1) is a stress response protein overexpressed upon cell injury in virtually all organs including the exocrine pancreas. Despite NUPR1's well-established role in the response to cell stress, the molecular and structural machineries triggered by NUPR1 activation remain largely debated. In this study, we uncover a new role for NUPR1, participating in the unfolded protein response (UPR) and the integrated stress response. Biochemical results and ultrastructural morphological observations revealed alterations in the UPR of acinar cells of germline-deleted NUPR1 murine models, consistent with the inability to restore general protein synthesis after stress induction. Bioinformatic analysis of NUPR1-interacting partners showed significant enrichment in translation initiation factors, including eukaryotic initiation factor (eIF) 2α. Co-immunoprecipitation and proximity ligation assays confirmed the interaction between NUPR1 and eIF2α and its phosphorylated form (p-eIF2α). Furthermore, our data suggest loss of NUPR1 in cells results in maintained eIF2α phosphorylation and evaluation of nascent proteins by click chemistry revealed that NUPR1-depleted PANC-1 cells displayed a slower poststress protein synthesis recovery when compared to wild-type. Combined, these data propose a novel role for NUPR1 in the integrated stress response pathway, at least partially through promoting efficient PERK branch activity and resolution through a unique interaction with eIF2α.
Collapse
Affiliation(s)
- Maria Teresa Borrello
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Marco Bocchio
- INMED (INSERM U1249), Turing Center for Living Systems, Aix-Marseille University, Marseille, France
| | - Angela Listi
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Nicolas Fraunhoffer
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Philippe Soubeyran
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Eric Chevet
- INSERM U1242, Proteostasis and Cancer Team, Chemistry Oncogenesis Stress Signaling, Université de Rennes 1, Rennes, France
| | - Christopher Pin
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
- Departments of Pediatrics, Oncology, and Physiology and Pharmacology, Schulich School of Medicine, University of Western Ontario, Children's Health Research Institute, London, ON, Canada
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
28
|
Li M. The role of P53 up-regulated modulator of apoptosis (PUMA) in ovarian development, cardiovascular and neurodegenerative diseases. Apoptosis 2021; 26:235-247. [PMID: 33783663 PMCID: PMC8197724 DOI: 10.1007/s10495-021-01667-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2021] [Indexed: 12/14/2022]
Abstract
P53 up-regulated modulator of apoptosis (PUMA), a pro-apoptotic BCL-2 homology 3 (BH3)-only member of the BCL-2 family, is a direct transcriptional target of P53 that elicits mitochondrial apoptosis under treatment with radiation and chemotherapy. It also induces excessive apoptosis in cardiovascular and/or neurodegenerative diseases. PUMA has been found to play a critical role in ovarian apoptosis. In the present paper, we review the progress of the study in PUMA over the past two decades in terms of its inducement and/or amplification of programmed cell death and describe recent updates to the understanding of both P53-dependent and P53-independent PUMA-mediated apoptotic pathways that are implicated in physiology and pathology, including the development of the ovary and cardiovascular and neurodegenerative diseases. We propose that PUMA may be a key regulator during ovary development, provide a model for PUMA-mediated apoptotic pathways, including intrinsic and extrinsic apoptotic pathways.
Collapse
Affiliation(s)
- Mei Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China.
| |
Collapse
|
29
|
Che J, Lv H, Yang J, Zhao B, Zhou S, Yu T, Shang P. Iron overload induces apoptosis of osteoblast cells via eliciting ER stress-mediated mitochondrial dysfunction and p-eIF2α/ATF4/CHOP pathway in vitro. Cell Signal 2021; 84:110024. [PMID: 33901579 DOI: 10.1016/j.cellsig.2021.110024] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/15/2022]
Abstract
Iron is an essential element for crucial biological function; whereas excess iron sedimentation impairs the main functions of tissues or organs. Cumulative researches have shown that the disturbances in iron metabolism, especially iron overload is closely concatenating with bone loss. Nevertheless, the specific process of iron overload-induced apoptosis in osteoblasts has not been thoroughly studied. In this study, our purpose is to elucidate the mechanism of osteoblast apoptosis induced by iron overload via the MC3T3-E1 cell line. Ferric ammonium citrate (FAC) was utilized to simulate iron overload conditions in vitro. These results showed that treatment with FAC dose-dependently induced the apoptosis of MC3T3-E1 cells at 48 h, dysfunction of iron metabolism, and increased intracellular reactive oxygen species (ROS) levels. Following, FAC does-dependently caused the calcium dyshomeostasis, decreased the calcium concentration in endoplasmic reticulum (ER), but increased the crosstalk between ER and mitochondria, and calcium concentration in the mitochondria. Moreover, FAC dose-dependently decreased mitochondrial membrane potential (MMP) and enhanced the expression of apoptosis related proteins (Bax, Cyto-C and C-caspase3). We furthermore revealed that FAC treatment activated the ER-mediated cell apoptosis via p-eIF2α/ATF4/CHOP pathway in MC3T3-E1 osteoblasts cells. In addition, pretreatment with the N-acetylcysteine (NAC) or Tauroursodeoxycholate Sodium (TUDC) attenuated cell apoptosis, ROS levels, mitochondria fragmentation and ER stress-related protein expression, and recovered the protein expression related to iron metabolism. In conclusion, our finding suggested that iron overload induced apoptosis via eliciting ER stress, which resulted in mitochondrial dysfunction and activated p-eIF2α/ATF4/CHOP pathway.
Collapse
Affiliation(s)
- Jingmin Che
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, Guangdong 518057, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Huanhuan Lv
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, Guangdong 518057, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Jiancheng Yang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, Guangdong 518057, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Department of Spinal Surgery, People's Hospital of Longhua Shenzhen, Shenzhen, China
| | - Bin Zhao
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, Guangdong 518057, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Sibo Zhou
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710072, China
| | - Tongyao Yu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, Guangdong 518057, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, Guangdong 518057, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| |
Collapse
|
30
|
Ko EJ, Ock MS, Choi YH, Iovanna JL, Mun S, Han K, Kim HS, Cha HJ. Human Endogenous Retrovirus (HERV)-K env Gene Knockout Affects Tumorigenic Characteristics of nupr1 Gene in DLD-1 Colorectal Cancer Cells. Int J Mol Sci 2021; 22:ijms22083941. [PMID: 33920455 PMCID: PMC8070087 DOI: 10.3390/ijms22083941] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 12/25/2022] Open
Abstract
Human endogenous retroviruses (HERVs) are suggested to be involved in the development of certain diseases, especially cancers. To elucidate the function of HERV-K Env protein in cancers, an HERV-K env gene knockout (KO) in DLD-1 colorectal cancer cell lines was generated using the CRISPR-Cas9 system. Transcriptome analysis of HERV-K env KO cells using next-generation sequencing (NGS) was performed to identify the key genes associated with the function of HERV-K Env protein. The proliferation of HERV-K env KO cells was significantly reduced in in vitro culture as well as in in vivo nude mouse model. Tumorigenic characteristics, including migration, invasion, and tumor colonization, were also significantly reduced in HERV-K env KO cells. Whereas, they were enhanced in HERV-K env over-expressing DLD-1 cells. The expression of nuclear protein-1 (NUPR1), an ER-stress response factor that plays an important role in cell proliferation, migration, and reactive oxygen species (ROS) generation in cancer cells, significantly reduced in HERV-K env KO cells. ROS levels and ROS-related gene expression was also significantly reduced in HERV-K env KO cells. Cells transfected with NUPR1 siRNA (small interfering RNA) exhibited the same phenotype as HERV-K env KO cells. These results suggest that the HERV-K env gene affects tumorigenic characteristics, including cell proliferation, migration, and tumor colonization through NUPR1 related pathway.
Collapse
Affiliation(s)
- Eun-Ji Ko
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea; (E.-J.K.); (M.-S.O.)
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 46241, Korea
| | - Mee-Sun Ock
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea; (E.-J.K.); (M.-S.O.)
| | - Yung-Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dongeui University, Busan 47227, Korea;
| | - Juan L. Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, France;
| | - Seyoung Mun
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Korea;
- Center for Bio-Medical Engineering Core Facility, Dankook University, Cheonan 31116, Korea;
| | - Kyudong Han
- Center for Bio-Medical Engineering Core Facility, Dankook University, Cheonan 31116, Korea;
- Department of Microbiology, Dankook University, Cheonan 31116, Korea
| | - Heui-Soo Kim
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 46241, Korea
- Correspondence: (H.-S.K.); (H.-J.C.); Tel.: +82-51-510-2259 (H.-S.K.); +82-51-990-6428 (H.-J.C.)
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea; (E.-J.K.); (M.-S.O.)
- Correspondence: (H.-S.K.); (H.-J.C.); Tel.: +82-51-510-2259 (H.-S.K.); +82-51-990-6428 (H.-J.C.)
| |
Collapse
|
31
|
Attenuating senescence and dead cells accumulation as heart failure therapy: Break the communication networks. Int J Cardiol 2021; 334:72-85. [PMID: 33794236 DOI: 10.1016/j.ijcard.2021.03.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 02/03/2023]
|
32
|
Tabatabaei Mirakabad FS, Khoramgah MS, Abdollahifar MA, Tehrani AS, Rezaei-Tavirani M, Niknazar S, Tahmasebinia F, Mahmoudiasl GR, Khoshsirat S, Abbaszadeh HA. NUPR1- CHOP experssion, autophagosome formation and apoptosis in the postmortem striatum of chronic methamphetamine user. J Chem Neuroanat 2021; 114:101942. [PMID: 33675952 DOI: 10.1016/j.jchemneu.2021.101942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/25/2021] [Accepted: 02/27/2021] [Indexed: 11/28/2022]
Abstract
Methamphetamine (Meth) is a neuro-stimulator substrate which might lead to neural cell death and the activation of several interconnected cellular pathways as well. However, the precise molecular mechanisms underlying Meth-induced neural cell death remained unclear yet. The current study aimed to assess the specific relationship between long-term Meth exposure and several endoplasmic reticulum stress, autophagy, and apoptosis associated markers including C/EBP homologous protein (CHOP), Tribbles homolog 3(Trib3), Nuclear protein 1(NUPR1), and Beclin-1 expression in postmortem human striatum. Therefore, the effects of long-term Meth exposure on autophagy and apoptosis in the striatum of postmortem users were evaluated and molecular, immunehistochemical, and histological examinations were performed on 10 control and 10 Meth-addicted brains. The level of CHOP, Trib3, NUPR1, and Beclin-1, Microtubule-associated proteins 1A/1B light chain 3B(LC3), Caspase 3, and Autophagy protein 5 (ATG5) were measured by using qPCR and immunohistochemistry. Stereological neural cell counting, Hematoxylin and Eosin, Nissl and Tunel staining were also performed. Based on our findings, the expression level of CHOP, Trib3, NUPR1, and Beclin-1 in the striatum of Meth group were significantly higher than the control group. Besides, the neuronal cell death was substantially increased in the striatum based on data obtained from the Tunel assay and the stereological analysis. Long-term presence of Meth in the brain can induce ER stress and overexpression of NUPR1 which is associated with the upregulation of CHOP, a pro-apoptotic transcription factor. Moreover, an increase in Trib3 expression is implicated in CHOP-dependent autophagic cell death during Meth-induced ER stress accompanied by an increase in neuronal cell death in the striatum of the postmortem human brains. Beclin 1 expression was also upregulated which may due to the activation of autophagic mechanisms upon prolonged Meth exposure.
Collapse
Affiliation(s)
- Fatemeh Sadat Tabatabaei Mirakabad
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Sadat Khoramgah
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Biology and Anatomical Sciencese, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefeh Shirazi Tehrani
- Department of Biology and Anatomical Sciencese, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Para Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Niknazar
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Shahrokh Khoshsirat
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Biology and Anatomical Sciencese, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Chen G, Wei X, Xu X, Yu G, Yong Z, Su R, Tao L. Methamphetamine Inhibits Long-Term Memory Acquisition and Synaptic Plasticity by Evoking Endoplasmic Reticulum Stress. Front Neurosci 2021; 14:630713. [PMID: 33519373 PMCID: PMC7840888 DOI: 10.3389/fnins.2020.630713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/14/2020] [Indexed: 01/23/2023] Open
Abstract
Methamphetamine (MA), an illicit drug abused worldwide, leads to cognitive impairment and memory loss. However, the detailed mechanisms of MA-induced neurologic impairment are still unclear. The present study aimed to investigate the mechanisms of MA-induced inhibition of memory acquisition from the perspective of endoplasmic reticulum (ER) stress. ER stress, caused by the accumulation of wrongly folded proteins in the ER, is important for new protein synthesis, which further influence the formation of long-term memory. A subacute MA poisoning model of mice was established and several behavioral experiments were performed, including elevated plus maze, Morris water maze, electro-stimulus Y-maze, and novel object recognition tasks. The present results suggested that 4 days exposure to MA induced significant memory loss. Whereas, this damage to memory formation could be protected when mice were pre-treated with ER stress inhibitor, tauroursodeoxycholic acid (TUDCA). The results of Western blotting showed that subacute exposure to MA increased the expression levels of ER stress marker proteins, such as binding immunoglobulin protein, phosphorylated eukaryotic translation initiation factor 2α, cyclic AMP-dependent transcription factor (ATF)-4, ATF-6, and CCAAT-enhancer binding protein homologous protein. Meanwhile, the enhanced expression levels of these proteins were reversed by TUDCA, indicating that MA administration induced memory loss by evoking ER stress in the hippocampus. We also found that MA inhibited the induction of long-term potentiation (LTP) in the hippocampus. Nevertheless, LTP could be induced when mice were pre-treated with TUDCA. In conclusion, MA inhibited long-term memory acquisition and synaptic plasticity via ER stress.
Collapse
Affiliation(s)
- Guang Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | | | - Xiang Xu
- School of Forensic Medicine, Wannan Medical College, Wuhu, China
| | - Gang Yu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zheng Yong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ruibin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Luyang Tao
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| |
Collapse
|
34
|
Mansour SMA, Ali SA, Nofal S, Soror SH. Targeting NUPR1 for Cancer Treatment: A Risky Endeavor. Curr Cancer Drug Targets 2020; 20:768-778. [PMID: 32619170 DOI: 10.2174/1568009620666200703152523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
NUPR1 is a transcription factor that has attracted great attention because of its various roles in cancer. Several studies were carried out to determine its molecular targets and mechanism of action to develop novel therapies against cancer. Here, we shed light on the role of NUPR1 in different types of cancer. NUPR1 regulates a complex network of pathways that may be affected by its silencing, which can cause varying effects. Its role in some types of cancer has been reported but remains incompletely understood, whereas its roles in other types of cancers have not been reported yet. Therefore, targeting NUPR1 for cancer treatment remains challenging and risky.
Collapse
Affiliation(s)
- Salma M A Mansour
- Egyptian Patent Office, Academy of Scientific Research and Technology (ASRT), 101 Kaser Al-Ainy Street, Cairo, Egypt
| | - Sahar A Ali
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Ain Helwan, Helwan Cairo 11795, Egypt
| | - Shaira Nofal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Ain Helwan, Helwan Cairo 11795, Egypt
| | - Sameh H Soror
- Egyptian Patent Office, Academy of Scientific Research and Technology (ASRT), 101 Kaser Al-Ainy Street, Cairo, Egypt.,Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Ain Helwan, Helwan Cairo 11795, Egypt
| |
Collapse
|
35
|
MicroRNA-325-3p prevents sevoflurane-induced learning and memory impairment by inhibiting Nupr1 and C/EBPβ/IGFBP5 signaling in rats. Aging (Albany NY) 2020; 12:5209-5220. [PMID: 32191629 PMCID: PMC7138556 DOI: 10.18632/aging.102942] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/27/2020] [Indexed: 12/17/2022]
Abstract
Endoplasmic reticulum stress-induced neuronal apoptosis contributes to neurotoxicity observed after sevoflurane exposure. However, the molecular mechanism underlying the resulting learning and memory impairments remains unknown. Here, we investigated the roles of miR-325-3p and Nupr1 in sevoflurane-induced learning and memory impairments in neonatal rats and HCN-2 human cortical neuronal cells. We found that in both neonatal rats and HCN-2 cells, sevoflurane exposure impairs learning and memory in neonatal rats and increases expression of Nupr1, the endoplasmic reticulum stress marker proteins C/EBPβ and IGFBP5, and the apoptosis-related protein markers cleaved-Caspase-3 and Bax. Using bioinformatics tools to identify microRNAs that bind to Nupr1, we found that miR-325-3p is downregulated in hippocampal neurons exposed to sevoflurane. Moreover, Nupr1 knockdown and miR-325-3p overexpression improved the rats’ performance in learning and memory tests and reduced sevoflurane-induced apoptosis in vitro and in vivo. These results suggest that miR-325-3p blocks sevoflurane-induced learning and memory impairments by inhibiting Nupr1 and the downstream C/EBPβ/IGFBP5 signaling axis in neonatal rats. MiR-325-3p may therefore be a useful therapeutic target in sevoflurane-induced neurotoxicity.
Collapse
|
36
|
Wu B, Zeng W, Ouyang W, Xu Q, Chen J, Wang B, Zhang X. Quercetin induced NUPR1-dependent autophagic cell death by disturbing reactive oxygen species homeostasis in osteosarcoma cells. J Clin Biochem Nutr 2020; 67:137-145. [PMID: 33041510 PMCID: PMC7533857 DOI: 10.3164/jcbn.19-121] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 12/25/2019] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma is a primary bone aggressive cancer, affecting adolescents worldwide. Quercetin (a natural polyphenolic compound) is a polyphenolic flavonoid compound found in a variety of plants. It has been demonstrated to exert cytostatic activity against a variety of human cancer, including the human osteosarcoma. However, its efficacy in the treatment of osteosarcoma and the underlying antitumor mechanism has not been fully elucidated yet. In this study, we exposed MG-63 cells to different concentrations of quercetin (50, 100 and 200 µM) for 24 h. Here, we show that quercetin increased autophagic flux in the MG-63 cells, as evidenced by the upregulation of LC3B-II/LC3B-I and downregulation of P62/SQSTM1. Moreover, the autophagy inhibitor Bafilomycin A1 or genetic blocking autophagy with ATG5 knockdown decreased quercetin-induced cell death, indicating quercetin triggered autophagic cell death in MG-63 cells. Specifically, quercetin increased NUPR1 expression and activated of NUPR1 reporter activity, which contributed to the expression of autophagy-related genes and subsequent initiated autophagic cell death in osteosarcoma cells. Importantly, the increased expression NUPR1 were tightly related to the disturbance of reactive oxygen species (ROS) homeostasis, which could be prevented by inhibiting intracellular ROS with NAC. Finally, NAC also abolished quercetin-induced autophagic cell death in vivo. Taken together, these data demonstrate that quercetin induces osteosarcoma cell death via inducing excessive autophagy, which is mediated through the ROS-NUPR1 pathway. Quercetin application may be a promising and practical strategy for osteosarcoma treatment in clinical practice.
Collapse
Affiliation(s)
- Bowen Wu
- Department of Orthopedics, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412007, China
| | - Wusi Zeng
- Department of Orthopedics, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412007, China
| | - Wei Ouyang
- Department of Oncology, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412007, China
| | - Qiang Xu
- Department of Orthopedics, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412007, China
| | - Jian Chen
- Department of Orthopedics, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412007, China
| | - Biao Wang
- Department of Orthopedics, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412007, China
| | - Xiping Zhang
- Department of Orthopedics, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412007, China
| |
Collapse
|
37
|
Li A, Li X, Chen X, Zeng C, Wang Z, Li Z, Chen J. NUPR1 Silencing Induces Autophagy-Mediated Apoptosis in Multiple Myeloma Cells Through the PI3K/AKT/mTOR Pathway. DNA Cell Biol 2020; 39:368-378. [PMID: 31971825 DOI: 10.1089/dna.2019.5196] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Anmao Li
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
| | - Xingxin Li
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
| | - Xuanxin Chen
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
| | - Chensi Zeng
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
| | - Zuo Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
| | - Zhen Li
- Department of Hematology, The First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Jianbin Chen
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
| |
Collapse
|
38
|
Huo W, Li H, Zhang Y, Li H. Epigenetic silencing of microRNA-874-3p implicates in erectile dysfunction in diabetic rats by activating the Nupr1/Chop-mediated pathway. FASEB J 2019; 34:1695-1709. [PMID: 31914690 DOI: 10.1096/fj.201902086r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
Abstract
Diabetes is a global medical problem that causes many deaths every year. Complications caused by diabetes are serious and affect patients' quality of life. Diabetes mellitus erectile dysfunction (DMED) affects more than half of male diabetes patients. In this study, we determined the role of microRNA-874-3p (miR-874-3p) and nuclear protein-1 (Nupr1) in streptozocin-induced DMED rats. Control rats received equal amount of vehicle. These rats were also injected with lentiviral vector or agomir to silence or overexpress miR-874-3p or Nupr1. Apomorphine (100 μg/kg, s.c.) was used to induce erection and time of erection was recorded. Intracavernosal and mean arterial pressure ratio (ICP/MAP) were also recorded. O2- level and concentration of thiobarbituric acid reactive substances (TBARs) were detected using lucigenin-derived chemiluminescence method and Colorimetry. Rat cavernosum tissues were collected for subsequent experiments. Cavernosum smooth muscle cells (CSMCs) were also used for in vitro experiments. Nupr1 was found highly expressed (by RT-qPCR and Western blot analysis) in cavernosum tissues from DMED rats. Nupr1 silencing improved the ICP/MAP ratio and erection time. Nupr1 silencing also reduced CSMC apoptosis (by TUNEL assay) as well as decreased O2- level and TBAR concentration. Nupr1 was targeted and inhibited by miR-874-3p (by luciferase activity and RNA immunoprecipitation assays), which was downregulated in DMED. miR-874-3p downregulation was due to increased methylation at the promoter region (methylation-specific PCR). miR-874-3p overexpression improved erection time and reduced apoptosis. In summary, miR-874-3p was downregulated which led to increased apoptosis and erectile dysfunction in DMED rats, through inhibition of Nupr1-mediated pathway. This study may also provide a new therapeutic direction for the treatment of DMED.
Collapse
Affiliation(s)
- Wei Huo
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Hongyan Li
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Yun Zhang
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Hai Li
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
39
|
R. Andrade P, Mehta M, Lu J, M. B. Teles R, Montoya D, O. Scumpia P, Nunes Sarno E, Ochoa MT, Ma F, Pellegrini M, Modlin RL. The cell fate regulator NUPR1 is induced by Mycobacterium leprae via type I interferon in human leprosy. PLoS Negl Trop Dis 2019; 13:e0007589. [PMID: 31344041 PMCID: PMC6684084 DOI: 10.1371/journal.pntd.0007589] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 08/06/2019] [Accepted: 06/30/2019] [Indexed: 11/18/2022] Open
Abstract
The initial interaction between a microbial pathogen and the host immune response influences the outcome of the battle between the host and the foreign invader. Leprosy, caused by the obligate intracellular pathogen Mycobacterium leprae, provides a model to study relevant human immune responses. Previous studies have adopted a targeted approach to investigate host response to M. leprae infection, focusing on the induction of specific molecules and pathways. By measuring the host transcriptome triggered by M. leprae infection of human macrophages, we were able to detect a host gene signature 24-48 hours after infection characterized by specific innate immune pathways involving the cell fate mechanisms autophagy and apoptosis. The top upstream regulator in the M. leprae-induced gene signature was NUPR1, which is found in the M. leprae-induced cell fate pathways. The induction of NUPR1 by M. leprae was dependent on the production of the type I interferon (IFN), IFN-β. Furthermore, NUPR1 mRNA and protein were upregulated in the skin lesions from patients with the multibacillary form of leprosy. Together, these data indicate that M. leprae induces a cell fate program which includes NUPR1 as part of the host response in the progressive form of leprosy.
Collapse
Affiliation(s)
- Priscila R. Andrade
- Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Manali Mehta
- Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Jing Lu
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Rosane M. B. Teles
- Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Dennis Montoya
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Phillip O. Scumpia
- Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | | | - Maria Teresa Ochoa
- Department of Dermatology, University of Southern California School of Medicine, Los Angeles, California, United States of America
| | - Feiyang Ma
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Robert L. Modlin
- Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
40
|
Fan T, Chen Y, He Z, Wang Q, Yang X, Ren Z, Zhang S. Inhibition of ROS/NUPR1-dependent autophagy antagonises repeated cadmium exposure -induced oral squamous cell carcinoma cell migration and invasion. Toxicol Lett 2019; 314:142-152. [PMID: 31319114 DOI: 10.1016/j.toxlet.2019.07.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/18/2019] [Accepted: 07/09/2019] [Indexed: 12/22/2022]
Abstract
Cadmium (Cd), an established carcinogen, is a risk factor for oral squamous cell carcinoma (OSCC). Macroautophagy/autophagy is proposed to play a pivotal role in Cd-mediated carcinogenic activity. However, the mechanisms underlying Cd-induced autophagy are poorly understood. In the present study, a CAL27 OSCC cell line exposed to 10-6 M Cd for 8 weeks was used as a model system. Repeated Cd exposure induced significant migration and invasion of CAL27 cells. Furthermore, we showed that Cd increased the autophagic flux in CAL27 cells, as evidenced by the upregulation of LC3-II and the downregulation of P62/SQSTM1. The genetic blocking of autophagy inhibited Cd-induced migration and invasion, indicating a carcinogenic role of autophagy in Cd-treated CAL27 cells. Cd-induced NUPR1 expression, which contributes to lysosomal biogenesis and expression of autophagy-related gene, was found to mechanistically initiate autophagy in CAL27 cells. Of note, NUPR1 shRNA abolished Cd-induced autophagy both in vitro and in vivo. We also found that Cd triggered the generation of MDA in a xenograft tumour model and that N-acetyl-l-cysteine, a reactive oxygen species (ROS) scavenger, abrogated the effects of Cd on NUPR1-dependent autophagy in vivo. Taken together, these results demonstrate that ROS-dependent NUPR1-mediated autophagy plays an important role in repeated Cd exposure -induced cell growth, migration and invasion in OSCC cells.
Collapse
Affiliation(s)
- Tengfei Fan
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yanrong Chen
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhijing He
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qing Wang
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xi Yang
- Department of Oral Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenhu Ren
- Department of Oral Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Sheng Zhang
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
41
|
Nupr1 regulates palmitate-induced apoptosis in human articular chondrocytes. Biosci Rep 2019; 39:BSR20181473. [PMID: 30674641 PMCID: PMC6379229 DOI: 10.1042/bsr20181473] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/04/2019] [Accepted: 01/09/2019] [Indexed: 12/18/2022] Open
Abstract
Obesity, a major risk factor for the development of osteoarthritis (OA), is associated with increased circulating levels of free fatty acids (FFA). However, the role of these FFAs in OA pathophysiology is not clearly understood. In the present study, we found that palmitate treatment of human primary articular chondrocytes increased the expression of ER stress markers [activating transcription factor 4 (ATF4), C/EBP homologous protein (CHOP)] and apoptosis markers [cytochrome c and cleaved caspase-3 (CC3)]. Palmitate treatment also increased the expression of Nuclear protein 1 (Nupr1) and tribbles related protein 3 (TRB3), which are known negative regulators of cell survival pathways. Knockdown of Nupr1 or CHOP expression inhibited palmitate mediated increased expression of TRB3 and CC3, indicating that Nupr1 and CHOP cooperate to regulate cell survival and apoptotic pathways in human chondrocytes. Nupr1 knockdown had no effect on CHOP expression whereas CHOP knockdown abolished the palmitate-mediated Nupr1 expression, indicating that CHOP is functional upstream to Nupr1 in this pathway. Moreover, overexpression of Nupr1 markedly increased the basal expression of pro-apoptotic molecules, including cytochrome c and CC3. Taken together, our study demonstrates that Nupr1 plays a crucial role in palmitate-induced apoptosis in human chondrocytes and Nupr1 as a potential novel drug target for the treatment of OA.
Collapse
|
42
|
Non-Structural Protein 2B of Human Rhinovirus 16 Activates Both PERK and ATF6 Rather Than IRE1 to Trigger ER Stress. Viruses 2019; 11:v11020133. [PMID: 30717233 PMCID: PMC6409610 DOI: 10.3390/v11020133] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/26/2019] [Accepted: 01/29/2019] [Indexed: 12/13/2022] Open
Abstract
To understand the underlying mechanisms of endoplasmic reticulum (ER) stress caused by human rhinovirus (HRV) 16 and non-structural transmembrane protein 2B, the expressions of ER chaperone glucose-regulated protein 78 (GRP78) and three signal transduction pathways, including protein kinase RNA-like ER kinase (PERK), activating transcription factor 6 (ATF6) and inositol-requiring enzyme 1 (IRE1), were evaluated after HRV16 infection and 2B gene transfection. Our results showed that both HRV16 infection and 2B gene transfection increased the expression of ER chaperone GRP78, and induced phosphorylation of PERK and cleavage of ATF6 in a time-dependent manner. Our data also revealed that the HRV16 2B protein was localized to the ER membrane. However, both HRV16 infection and HRV16 2B gene transfection did not induce ER stress through the IRE1 pathway. Moreover, our results showed that apoptosis occurred in H1-HeLa cells infected with HRV16 or transfected with 2B gene accompanied with increased expression of CHOP and cleaved caspase-3. Taken together, non-structural protein 2B of HRV16 induced an ER stress response through the PERK and ATF6 pathways rather than the IRE1 pathway.
Collapse
|
43
|
Du SH, Zhang W, Yue X, Luo XQ, Tan XH, Liu C, Qiao DF, Wang H. Role of CXCR1 and Interleukin-8 in Methamphetamine-Induced Neuronal Apoptosis. Front Cell Neurosci 2018; 12:230. [PMID: 30123110 PMCID: PMC6085841 DOI: 10.3389/fncel.2018.00230] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022] Open
Abstract
Methamphetamine (METH), an extremely and widely abused illicit drug, can cause serious nervous system damage and social problems. Previous research has shown that METH use causes dopaminergic neuron apoptosis and astrocyte-related neuroinflammation. However, the relationship of astrocytes and neurons in METH-induced neurotoxicity remains unclear. We hypothesized that chemokine interleukin (IL) eight released by astrocytes and C-X-C motif chemokine receptor 1 (CXCR1) in neurons are involved in METH-induced neuronal apoptosis. We tested our hypothesis by examining the changes of CXCR1 in SH-SY5Y cells and in the brain of C57BL/6 mice exposed to METH by western blotting and immunolabeling. We also determined the effects of knocking down CXCR1 expression with small interfering ribonucleic acid (siRNA) on METH-exposed SH-SY5Y cells. Furthermore, we detected the expression levels of IL-8 and the nuclear factor-kappa B (NF-κB) pathway in U87MG cells and then co-cultured the two cell types to determine the role of CXCR1 and IL-8 in neuronal apoptosis. Our results indicated that METH exposure increased CXCR1 expression both in vitro and in vivo, with the effects obtained in vitro being dose-dependent. Silencing of CXCR1 expression with siRNAs reduced the expression of cleaved caspase-3, cleaved poly (ADP-ribose) polymerase (PARP), and other related proteins. In addition, IL-8 expression and release were increased in METH-exposed U87MG cells, which is regulated by NF-κB pathway. Neuronal apoptosis was attenuated by siCXCR1 after METH treatment in the co-cultured cells, which can be reversed after exposure to recombinant IL-8. These results demonstrate that CXCR1 plays an important role in neuronal apoptosis induced by METH and may be a potential target for METH-induced neurotoxicity therapy. Highlights -Methamphetamine exposure upregulated the expression of CXCR1.-Methamphetamine exposure increased the expression of interleukin-8 through nuclear factor-kappa B pathway.-Activation of CXCR1 by interleukin-8 induces an increase in methamphetamine-related neuronal apoptosis.
Collapse
Affiliation(s)
- Si-Hao Du
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Wei Zhang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xia Yue
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xiao-Qing Luo
- School of Forensic Medicine, Southern Medical University, Guangzhou, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-Hui Tan
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Chao Liu
- Guangzhou Forensic Science Institute, Guangzhou Public Security Bureau, Guangzhou, China
| | - Dong-Fang Qiao
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
44
|
Xu X, Huang E, Luo B, Cai D, Zhao X, Luo Q, Jin Y, Chen L, Wang Q, Liu C, Lin Z, Xie WB, Wang H. Methamphetamine exposure triggers apoptosis and autophagy in neuronal cells by activating the C/EBPβ-related signaling pathway. FASEB J 2018; 32:fj201701460RRR. [PMID: 29939784 DOI: 10.1096/fj.201701460rrr] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Methamphetamine (Meth) is a widely abused psychoactive drug that primarily damages the nervous system, notably causing dopaminergic neuronal apoptosis. CCAAT-enhancer binding protein (C/EBPβ) is a transcription factor and an important regulator of cell apoptosis and autophagy. Insulin-like growth factor binding protein (IGFBP5) is a proapoptotic factor that mediates Meth-induced neuronal apoptosis, and Trib3 (tribbles pseudokinase 3) is an endoplasmic reticulum (ER) stress-inducible gene involved in autophagic cell death through the mammalian target of rapamycin (mTOR) signaling pathway. To test the hypothesis that C/EBPβ is involved in Meth-induced IGFBP5-mediated neuronal apoptosis and Trib3-mediated neuronal autophagy, we measured the protein expression of C/EBPβ after Meth exposure and evaluated the effects of silencing C/EBPβ, IGFBP5, or Trib3 on Meth-induced apoptosis and autophagy in neuronal cells and in the rat striatum after intrastriatal Meth injection. We found that, at relatively high doses, Meth exposure increased C/EBPβ protein expression, which was accompanied by increased neuronal apoptosis and autophagy; triggered the IGFBP5-mediated, p53-up-regulated modulator of apoptosis (PUMA)-related mitochondrial apoptotic signaling pathway; and stimulated the Trib3-mediated ER stress signaling pathway through the Akt-mTOR signaling axis. We also found that autophagy is an early response to Meth-induced stress upstream of apoptosis and plays a detrimental role in Meth-induced neuronal cell death. These results suggest that Meth exposure induces C/EBPβ expression, which plays an essential role in the neuronal apoptosis and autophagy induced by relatively high doses of Meth; however, relatively low concentrations of Meth did not change the expression of C/EBPβ in vitro. Further studies are needed to elucidate the role of C/EBPβ in low-dose Meth-induced neurotoxicity.-Xu, X., Huang, E., Luo, B., Cai, D., Zhao, X., Luo, Q., Jin, Y., Chen, L., Wang, Q., Liu, C., Lin, Z., Xie, W.-B., Wang, H. Methamphetamine exposure triggers apoptosis and autophagy in neuronal cells by activating the C/EBPβ-related signaling pathway.
Collapse
Affiliation(s)
- Xiang Xu
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
- School of Forensic Medicine, Wannan Medical College, Wuhu, China
| | - Enping Huang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Baoying Luo
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Dunpeng Cai
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xu Zhao
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Qin Luo
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Yili Jin
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Ling Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Qi Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Chao Liu
- Guangzhou Forensic Science Institute, Guangzhou, China; and
| | - Zhoumeng Lin
- Department of Anatomy and Physiology, Institute of Computational Comparative Medicine, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Wei-Bing Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
45
|
Zhao C, Mei Y, Chen X, Jiang L, Jiang Y, Song X, Xiao H, Zhang J, Wang J. Autophagy plays a pro-survival role against methamphetamine-induced apoptosis in H9C2 cells. Toxicol Lett 2018; 294:156-165. [PMID: 29763685 DOI: 10.1016/j.toxlet.2018.05.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 03/03/2018] [Accepted: 05/11/2018] [Indexed: 01/24/2023]
Abstract
Methamphetamine (METH) is a commonly abused psychostimulant that can induce severe neurotoxicity. Cardiovascular injury caused by METH has recently gained increasing attention; however, the underlying mechanisms remain unclear. As autophagy has been shown to be associated with cell injury, the association between autophagy and METH-mediated cell apoptosis was investigated in the present study. METH treatment significantly increased the expression of two key autophagy proteins, i.e., Beclin-1 and LC3-II, in the cardiomyocyte cell line H9C2. Furthermore, according to western blot and flow cytometry analyses, METH contributed to cell injury and markedly enhanced cleaved-caspase 3 and PARP expression. In addition, the corresponding AKT-mTOR survival pathway axis was appeared deactivated. The autophagic activity was closely associated with METH-mediated cell injury because rapamycin, which is an autophagy inducer, markedly attenuated METH-induced cell injury, while 3-Methyladenine (3-MA), which is an autophagy inhibitor, and bafilomycinA1 (Baf-A1), which is a blocker of autophagosome-lysosome fusion, markedly exacerbated METH-induced cell injury. Notably, defective autophagosome-lysosome fusion might be partially involved in the METH-induced enhancement of LC3-II expression and cell injury. However, the underlying mechanisms require further investigation.
Collapse
Affiliation(s)
- Chao Zhao
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Yong Mei
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Xufeng Chen
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Lei Jiang
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Yunfei Jiang
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Xu Song
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tian Yuan East Road, Nanjing, Jiangsu, 211166, China
| | - Hang Xiao
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tian Yuan East Road, Nanjing, Jiangsu, 211166, China
| | - Jingsong Zhang
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China.
| | - Jun Wang
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tian Yuan East Road, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
46
|
Nazari A, Zahmatkesh M, Mortaz E, Hosseinzadeh S. Effect of methamphetamine exposure on the plasma levels of endothelial-derived microparticles. Drug Alcohol Depend 2018; 186:219-225. [PMID: 29609134 DOI: 10.1016/j.drugalcdep.2018.02.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/28/2018] [Accepted: 02/01/2018] [Indexed: 11/15/2022]
Abstract
BACKGROUND Methamphetamine (Meth), a neurotoxin, induces inflammation, oxidative stress, and triggers endothelial dysfunction and cardiovascular disease which is the second cause of death among individuals with Meth-use disorder. Oxidative stress and inflammation trigger the microparticle (MP) release. These are extracellular vesicles extracted from cell surface and identified in biological fluids. MP levels alter during pathological conditions, suggesting its potential biomarker role. In this respect, we designed the present experiment to investigate the effects of Meth on the plasma level of the endothelial-derived microparticle (EMP). METHODS Animals received Meth (4 mg/kg i.p.) for 1, 7 and 14 days and then, the plasma level of EMPs was evaluated, using cell surface markers, including AnnexinV, CD144, CD31, CD41a antigens with the flow cytometry method. The biochemical indices and locomotor activity were also assessed in a rat model. RESULTS Meth increased locomotor activity (Meth-1, 277.12 ± 20.17; Meth-7, 262.25 ± 11.95; Meth-14, 265.75 ± 14.75), inflammatory and oxidative indices as evidenced by rising of the C-reactive protein (Meth-7, 39.4 ± 1.24; Meth-14, 38.58 ± 2.19, vs 8.65 ± 0.45, mg/L) and malondialdehyde (Meth-7, 9.74 ± 1.38; Meth-14, 14.6 ± 1.45, vs 4.43 ± 0.32 nmol/L) plasma levels. We also found that Meth triggered endothelial injury, as demonstrated by elevated levels of EMP (Meth-7, 4.77 ± 0.22; Meth-14, 5.91 ± 0.34, % total events/mL) compared with control group. CONCLUSION Our data showed that Meth exposure stimulates inflammatory and oxidative pathways and facilitates the EMPs shedding. Measuring the level of EMPs might be applied as a potential diagnostic index to monitor the endothelial dysfunction in substance-use disorders.
Collapse
Affiliation(s)
- Azadeh Nazari
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Zahmatkesh
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| | - Esmaeil Mortaz
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands; Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran- Iran
| | - Soheila Hosseinzadeh
- Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
47
|
Li L, Chen S, Wang Y, Yue X, Xu J, Xie W, Qiu P, Liu C, Wang A, Wang H. Role of GSK3β/α-synuclein axis in methamphetamine-induced neurotoxicity in PC12 cells. Toxicol Res (Camb) 2018; 7:221-234. [PMID: 30090577 PMCID: PMC6062219 DOI: 10.1039/c7tx00189d] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 12/22/2017] [Indexed: 12/22/2022] Open
Abstract
Methamphetamine (METH) is well-known as a potent psychostimulant of abuse worldwide. METH administration can cause neurotoxicity and neurodegenerative injury, which are similar to the two prevalent neurodegenerative disorders Alzheimer's disease (AD) and Parkinson's disease (PD). Recent results suggested that METH exposure increased the level of α-synuclein (α-syn) that could be a possible cause of neurotoxicity. However, the mechanism of METH-induced neurodegeneration remains unclear. This study was aimed at examining the effects of glycogen synthase kinase3β (GSK3β), α-syn, and tau on METH-induced neurotoxicity. Our results indicated that P-GSK3β (Tyr216), P-Tau (Ser396), α-syn, and P-α-syn (Ser129) levels were increased after METH administration in dose- and time-dependent manners. Upon inhibiting the GSK3β activity with LiCl or GSK3β-siRNA, these protein expressions were significantly decreased. We observed that LiCl protected the cells from METH-caused cytotoxicity by weakening the cell morphological damage and preventing cell apoptosis and death. We also found that P-GSK3β colocalized with P-Tau and α-syn by the immunofluorescence method. Further, METH disrupted the cellular autophagy by upregulation of LC3-II and P62 proteins, and the cellular autophagy was restored by LiCl and GSK3β-siRNA. The expressions of the α-syn-specific degradative enzyme glucocerebrosidase (GCase) with its regulator lysosomal integral membrane protein type-2 (LIMP-2) decreased inversely with the doses of METH treatment. The GCase inhibitor conduritol-β-epoxide (CβE) increased the α-syn levels, and LiCl restored GCase and LIMP-2 expressions disrupted by the METH treatment. In summary, we conclude that GSK3β plays key roles in METH-induced neurotoxicity and neurodegenerative injury by promoting abnormal protein phosphorylation and α-syn accumulation, blocking the autophagy-lysosomal degradation pathway, and finally leading to cell apoptosis and death. GSK3β may be a potential target to prevent METH-induced neurodegeneration.
Collapse
Affiliation(s)
- Lizeng Li
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Si Chen
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Yue Wang
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Xia Yue
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Jingtao Xu
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Weibing Xie
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Pingming Qiu
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Chao Liu
- Guangzhou Forensic Science Institute , Guangzhou 510030 , People's Republic of China
| | - AiFeng Wang
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Huijun Wang
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| |
Collapse
|
48
|
Du SH, Qiao DF, Chen CX, Chen S, Liu C, Lin Z, Wang H, Xie WB. Toll-Like Receptor 4 Mediates Methamphetamine-Induced Neuroinflammation through Caspase-11 Signaling Pathway in Astrocytes. Front Mol Neurosci 2017; 10:409. [PMID: 29311802 PMCID: PMC5733023 DOI: 10.3389/fnmol.2017.00409] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/27/2017] [Indexed: 02/02/2023] Open
Abstract
Methamphetamine (METH) is an amphetamine-typed stimulant drug that is increasingly being abused worldwide. Previous studies have shown that METH toxicity is systemic, especially targeting dopaminergic neurons in the central nervous system (CNS). However, the role of neuroinflammation in METH neurotoxicity remains unclear. We hypothesized that Toll-like receptor 4 (TLR4) and Caspase-11 are involved in METH-induced astrocyte-related neuroinflammation. We tested our hypothesis by examining the changes of TLR4 and Caspase-11 protein expression in primary cultured C57BL/6 mouse astrocytes and in the midbrain and striatum of mice exposed to METH with western blot and double immunofluorescence labeling. We also determined the effects of blocking Caspase-11 expression with wedelolactone (a specific inhibitor of Caspase-11) or siRNA on METH-induced neuroinflammation in astrocytes. Furthermore, we determined the effects of blocking TLR4 expression with TAK-242 (a specific inhibitor of TLR4) or siRNA on METH-induced neuroinflammation in astrocytes. METH exposure increased Caspase-11 and TLR4 expression both in vitro and in vivo, with the effects in vitro being dose-dependent. Inhibition of Caspase-11 expression with either wedelolactone or siRNAs reduced the expression of inflammasome NLRP3 and pro-inflammatory cytokines. In addition, blocking TLR4 expression inhibited METH-induced activation of NF-κB and Caspase-11 in vitro and in vivo, suggesting that TLR4-Caspase-11 pathway is involved in METH-induced neuroinflammation. These results indicate that Caspase-11 and TLR4 play an important role in METH-induced neuroinflammation and may be potential gene targets for therapeutics in METH-caused neurotoxicity.
Collapse
Affiliation(s)
- Si-Hao Du
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Dong-Fang Qiao
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Chuan-Xiang Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Si Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Chao Liu
- Guangzhou Forensic Science Institute, Guangzhou, China
| | - Zhoumeng Lin
- Department of Anatomy and Physiology, Institute of Computational Comparative Medicine (ICCM), College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Wei-Bing Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
49
|
Qie X, Wen D, Guo H, Xu G, Liu S, Shen Q, Liu Y, Zhang W, Cong B, Ma C. Endoplasmic Reticulum Stress Mediates Methamphetamine-Induced Blood-Brain Barrier Damage. Front Pharmacol 2017; 8:639. [PMID: 28959203 PMCID: PMC5603670 DOI: 10.3389/fphar.2017.00639] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/29/2017] [Indexed: 12/22/2022] Open
Abstract
Methamphetamine (METH) abuse causes serious health problems worldwide, and long-term use of METH disrupts the blood-brain barrier (BBB). Herein, we explored the potential mechanism of endoplasmic reticulum (ER) stress in METH-induced BBB endothelial cell damage in vitro and the therapeutic potential of endoplasmic reticulum stress inhibitors for METH-induced BBB disruption in C57BL/6J mice. Exposure of immortalized BMVEC (bEnd.3) cells to METH significantly decreased cell viability, induced apoptosis, and diminished the tightness of cell monolayers. METH activated ER stress sensor proteins, including PERK, ATF6, and IRE1, and upregulated the pro-apoptotic protein CHOP. The ER stress inhibitors significantly blocked the upregulation of CHOP. Knockdown of CHOP protected bEnd.3 cells from METH-induced cytotoxicity. Furthermore, METH elevated the production of reactive oxygen species (ROS) and induced the dysfunction of mitochondrial characterized by a Bcl2/Bax ratio decrease, mitochondrial membrane potential collapse, and cytochrome c. ER stress release was partially reversed by ROS inhibition, and cytochrome c release was partially blocked by knockdown of CHOP. Finally, PBA significantly attenuated METH-induced sodium fluorescein (NaFluo) and Evans Blue leakage, as well as tight junction protein loss, in C57BL/6J mice. These data suggest that BBB endothelial cell damage was caused by METH-induced endoplasmic reticulum stress, which further induced mitochondrial dysfunction, and that PBA was an effective treatment for METH-induced BBB disruption.
Collapse
Affiliation(s)
- Xiaojuan Qie
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China.,Department of Anesthesiology, The Third Hospital of Hebei Medical UniversityShijiazhuang, China
| | - Di Wen
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| | - Hongyan Guo
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| | - Guanjie Xu
- Department of Anesthesiology, The Third Hospital of Hebei Medical UniversityShijiazhuang, China
| | - Shuai Liu
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| | - Qianchao Shen
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| | - Yi Liu
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| | - Wenfang Zhang
- The 8th Brigade of General Division of Criminal Investigation, Beijing Municipal Public Security BureauBeijing, China
| | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| | - Chunling Ma
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| |
Collapse
|
50
|
Zhang Y, Zhang Y, Bai Y, Chao J, Hu G, Chen X, Yao H. Involvement of PUMA in pericyte migration induced by methamphetamine. Exp Cell Res 2017; 356:28-39. [PMID: 28408317 DOI: 10.1016/j.yexcr.2017.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 12/21/2022]
Abstract
Mounting evidence indicates that methamphetamine causes blood-brain barrier damage, with emphasis on endothelial cells. The role of pericytes in methamphetamine-induced BBB damage remains unknown. Our study demonstrated that methamphetamine increased the migration of pericytes from the endothelial basement membrane. However, the detailed mechanisms underlying this process remain poorly understood. Thus, we examined the molecular mechanisms involved in methamphetamine-induced pericyte migration. The results showed that exposure of C3H/10T1/2 cells and HBVPs to methamphetamine increased PUMA expression via activation of the sigma-1 receptor, MAPK and Akt/PI3K pathways. Moreover, methamphetamine treatment resulted in the increased migration of C3H/10T1/2 cells and HBVPs. Knockdown of PUMA in pericytes transduced with PUMA siRNA attenuated the methamphetamine-induced increase in cell migration through attenuation of integrin and tyrosine kinase mechanisms, implicating a role of PUMA in the migration of C3H/10T1/2 cells and HBVPs. This study has demonstrated that methamphetamine-mediated pericytes migration involves PUMA up-regulation. Thus, targeted studies of PUMA could provide insights to facilitate the development of a potential therapeutic approach for alleviation of methamphetamine-induced pericyte migration.
Collapse
Affiliation(s)
- Yanhong Zhang
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China.
| | - Yuan Zhang
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China.
| | - Ying Bai
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China.
| | - Jie Chao
- Department of Physiology, Medical School of Southeast University, Nanjing, China.
| | - Gang Hu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China.
| | - Xufeng Chen
- Department of Emergency, Jiangsu Province Hospital, Nanjing, Jiangsu, China.
| | - Honghong Yao
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China; Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|