1
|
Lafuente JV, Sharma A, Feng L, Muresanu DF, Nozari A, Tian ZR, Buzoianu AD, Sjöquist PO, Wiklund L, Sharma HS. Nanowired Delivery of Mesenchymal Stem Cells with Antioxidant Compound H-290/51 Reduces Exacerbation of Methamphetamine Neurotoxicity in Hot Environment. ADVANCES IN NEUROBIOLOGY 2023; 32:317-352. [PMID: 37480465 DOI: 10.1007/978-3-031-32997-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Military personnel are often exposed to hot environments either for combat operations or peacekeeping missions. Hot environment is a severe stressful situation leading to profound hyperthermia, fatigue and neurological impairments. To avoid stressful environment, some people frequently use methamphetamine (METH) or other psychostimulants to feel comfortable under adverse situations. Our studies show that heat stress alone induces breakdown of the blood-brain barrier (BBB) and edema formation associated with reduced cerebral blood flow (CBF). On the other hand, METH alone induces hyperthermia and neurotoxicity. These effects of METH are exacerbated at high ambient temperatures as seen with greater breakdown of the BBB and brain pathology. Thus, a combination of METH use at hot environment may further enhance the brain damage-associated behavioral dysfunctions. METH is well known to induce severe oxidative stress leading to brain pathology. In this investigation, METH intoxication at hot environment was examined on brain pathology and to explore suitable strategies to induce neuroprotection. Accordingly, TiO2-nanowired delivery of H-290/51 (150 mg/kg, i.p.), a potent chain-breaking antioxidant in combination with mesenchymal stem cells (MSCs), is investigated in attenuating METH-induced brain damage at hot environment in model experiments. Our results show that nanodelivery of H-290/51 with MSCs significantly enhanced CBF and reduced BBB breakdown, edema formation and brain pathology following METH exposure at hot environment. These observations are the first to point out that METH exacerbated brain pathology at hot environment probably due to enhanced oxidative stress, and MSCs attenuate these adverse effects, not reported earlier.
Collapse
Affiliation(s)
- José Vicente Lafuente
- LaNCE, Department Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan, Hebei Province, China
| | - Dafin F Muresanu
- Department Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Ala Nozari
- Anesthesiology & Intensive Care, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Z Ryan Tian
- Department Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Per-Ove Sjöquist
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
2
|
Zeng Y, Chen Y, Zhang S, Ren H, Xia J, Liu M, Shan B, Ren Y. Natural Products in Modulating Methamphetamine-Induced Neuronal Apoptosis. Front Pharmacol 2022; 12:805991. [PMID: 35058785 PMCID: PMC8764133 DOI: 10.3389/fphar.2021.805991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Methamphetamine (METH), an amphetamine-type psychostimulant, is highly abused worldwide. Chronic abuse of METH causes neurodegenerative changes in central dopaminergic neurons with numerous neuropsychiatric consequences. Neuronal apoptosis plays a critical role in METH-induced neurotoxicity and may provide promising pharmacological targets for preventing and treating METH addiction. In recent years, accumulating evidence has revealed that natural products may possess significant potentials to inhibit METH-evoked neuronal apoptosis. In this review, we summarized and analyzed the improvement effect of natural products on METH-induced neuronal apoptosis and their potential molecular mechanisms on modulating dopamine release, oxidative stress, mitochondrial-dependent apoptotic pathway, endoplasmic reticulum stress-mediated apoptotic pathway, and neuroinflammation. Hopefully, this review may highlight the potential value of natural products in modulating METH-caused neuronal apoptosis and provide useful information for future research and developments of novel and efficacious pharmacotherapies in this field.
Collapse
Affiliation(s)
- Yiwei Zeng
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunhui Chen
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Su Zhang
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Ren
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jialin Xia
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengnan Liu
- Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, China
| | - Baozhi Shan
- School of Humanities, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yulan Ren
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
3
|
Ghafoor H, Chu H, Huang J, Chen M, Wang S, Wang J, Chao J. ZC3H4 promotes pulmonary fibrosis via an ER stress-related positive feedback loop. Toxicol Appl Pharmacol 2022; 435:115856. [PMID: 34979141 DOI: 10.1016/j.taap.2021.115856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Pulmonary fibrosis is a sequela of many pulmonary diseases, such as pneumoconiosis and idiopathic pulmonary fibrosis. The principal characteristics of pulmonary fibrosis comprise myofibroblast proliferation, alveolar damage and deposition of extracellular matrix components, which cause abnormal lung structure remodeling and an irreversible decline in lung function; however, the detailed mechanisms remain unclear. The current study focused on the role of ZC3H4, a new member of the zinc finger protein family, in SiO2-induced pulmonary fibrosis. METHODS The expression of ZC3H4 and fibroblast activation markers (COL1A1, COL3A1 and ACTA1) was measured by western blotting and immunofluorescence staining after SiO2 exposure (50 μg/cm2). The functional change in fibroblasts was studied with a scratch assay and a 3D migration assay. The CRISPR/Cas9 system was used to explore the regulatory mechanisms of ZC3H4 in pulmonary fibroblast cells. RESULTS The expression levels of ZC3H4 and sigmar1 (a key regulator of ER stress) were increased in pulmonary fibroblast cells and were associated with fibroblast activation, as indicated by the increase in COL1A1, COL3A1 and ACTA1, as well as the migration ability. SiO2-enhanced fibroblast activation was attenuated by specific knockdown of ZC3H4 and inhibition of ER stress, demonstrating that ZC3H4 activated fibroblasts via the sigmar1/ER stress pathway. Interestingly, ER stress blockade also inhibited ZC3H4 expression, indicating the positive feedback regulatory mechanism of ER stress on ZC3H4. CONCLUSIONS Our results demonstrate that ZC3H4 and sigmar1 might act as novel therapeutic targets for silicosis, providing a reference for further pulmonary fibrosis research.
Collapse
Affiliation(s)
- Hammad Ghafoor
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 2100096, China; Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu 2100096, China; Department of Microbiology and Immunology, School of Medicine, Southeast University, Nanjing, Jiangsu 2100096, China; Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Han Chu
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 2100096, China; Key Laboratory of Development Genes and Human Disease, Southeast University, Nanjing, Jiangsu 2100096, China
| | - Jie Huang
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 2100096, China
| | - Mengling Chen
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 2100096, China
| | - Sha Wang
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 2100096, China
| | - Jing Wang
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 2100096, China.
| | - Jie Chao
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 2100096, China; Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Development Genes and Human Disease, Southeast University, Nanjing, Jiangsu 2100096, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; School of Medicine, Xizang Minzu University, Xianyang, Shanxi 712082, China.
| |
Collapse
|
4
|
Recombinant osteopontin provides protection for cerebral infarction by inhibiting the NLRP3 inflammasome in microglia. Brain Res 2020; 1751:147170. [PMID: 33157099 DOI: 10.1016/j.brainres.2020.147170] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 12/25/2022]
Abstract
Neuroinflammation is one of the most important secondary pathological events after cerebral infarction. Activation of NLRP3 inflammasome is a pivotal form of neuroinflammation. Osteopontin (OPN) is expressed during the subacute phase after cerebral infarction and has an important chemotactic effect on microglia. The aim of this study was to reveal the effect of recombinant OPN on brain injury after cerebral infarction and the regulation of NLRP3 inflammasome. We used the middle cerebral artery occlusion (MCAO) method-established focal cerebral ischemia model and LPS-induced inflammation model on neonate rat primary microglia. The effects of OPN on cerebral ischemic injury, neural function, microglia inflammation and NLRP3 inflammasome function were studied by immunofluorescence, staining, enzyme-linked immunosorbent assay and Western blot assay. We established MCAO cerebral ischemia and reperfusion injury model, and found that recombinant OPN reduced the volume of cerebral infarction and alleviated the ischemic injury degree of cerebral tissues, neurons, and neurological function. We found that OPN was also involved in the negative regulation of inflammasome and microglia activity in cerebral ischemic injury, and that OPN inhibited the activation of NLRP3 inflammasome and the function of microglia in a LPS-induced inflammatory model. Our findings show that recombinant OPN can reduce the ischemic infarct size and alleviate the cerebral ischemic injury of rats, which may be related to its efficient involvement in the inhibitory regulation of inflammasome and microglia inflammatory activation.
Collapse
|
5
|
Li J, Sun Q, Zhu S, Xi K, Shi Q, Pang K, Liu X, Li M, Zhang Y, Sun J. Knockdown of circHomer1 ameliorates METH-induced neuronal injury through inhibiting Bbc3 expression. Neurosci Lett 2020; 732:135050. [PMID: 32450188 DOI: 10.1016/j.neulet.2020.135050] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/25/2020] [Accepted: 05/10/2020] [Indexed: 12/19/2022]
Abstract
Current studies have illustrated that circular RNAs (circRNAs) are a vital part of non-coding RNA (ncRNAs) species and highly abundant and dynamically expressed in brain. However, the exact mechanisms by which circRNAs modulate methamphetamine (METH)-induced neuronal damage still remain largely unexplored. Consistent with our previous study, the expression of circHomer1 was significantly up-regulated after METH treatment in HT-22 cells. We confirmed its loop structure by detection of its back-splice junction with qRT-PCR product via sequence. Moreover, circHomer1 was resistant against RNase R digestion compared with its linear mRNA Homer1. Inhibition of circHomer1 expression indeed alleviated METH-induced neurotoxicity, with lower apoptosis rate via flow cytometry and cleaved Caspase3 protein level. Furthermore, we speculated that Bbc3 functioned as a target of circHomer1 based on computational algorithm, and knockdown of circHomer1 actually reduced Bbc3 expression at the mRNA and protein level. Besides, suppression of Bbc3 decreased the reactive oxygen species (ROS) level and radio of PI-positive cells. Furthermore, we analyzed the correlation in pairs among circHomer1, Bbc3 and behaviors in well-developed METH-addicted models using Pearson's correlation coefficient, which implied an important role of circHomer1 and Bbc3 in addictive behaviors. In all, we for the first time identified a novel circRNA, circHomer1 and our results suggested that circHomer1 regulated METH-induced lethal process by suppressing the Bbc3 expression.
Collapse
Affiliation(s)
- Junwei Li
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Qiyun Sun
- Department of Orthopedics, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, 277100, China
| | - Shaowei Zhu
- Department of Neurology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Kaiyan Xi
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Qingqing Shi
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Kunkun Pang
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Xiaoyu Liu
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Meng Li
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Yue Zhang
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Jinhao Sun
- Department of Anatomy, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
6
|
Levo-tetrahydropalmatine attenuates methamphetamine reward behavior and the accompanying activation of ERK phosphorylation in mice. Neurosci Lett 2020; 714:134416. [DOI: 10.1016/j.neulet.2019.134416] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/20/2019] [Accepted: 08/05/2019] [Indexed: 01/23/2023]
|