1
|
Maltaneri RE, Chamorro ME, Gionco SE, Nesse AB, Vittori DC. Erythropoietin enhances iron bioavailability in HepG2 cells by downregulating hepcidin through mTOR, C/EBPα and HIF-1α. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119800. [PMID: 39047915 DOI: 10.1016/j.bbamcr.2024.119800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
The regulation of iron (Fe) levels is essential to maintain an adequate supply for erythropoiesis, among other processes, and to avoid possible toxicity. The liver-produced peptide hepcidin is regarded as the main regulator of Fe absorption in enterocytes and release from hepatocytes and macrophages, as it impairs Fe export through ferroportin. The glycoprotein erythropoietin (Epo) drives erythroid progenitor survival and differentiation in the bone marrow, and has been linked to the mobilization of Fe reserves necessary for hemoglobin production. Herein we show that Epo inhibits hepcidin expression directly in the HepG2 hepatic cell line, thus leading to a decrease in intracellular Fe levels. Such inhibition was dependent on the Epo receptor-associated kinase JAK2, as well as on the PI3K/AKT/mTOR pathway, which regulates nutrient homeostasis. Epo was also found to decrease binding of the C/EBP-α transcription factor to the hepcidin promoter, which could be attributed to an increased expression of its inhibitor CHOP. Epo did not only hinder the stimulating effect of C/EBP-α on hepcidin transcription, but also favored hepcidin inhibition by HIF-1α, by increasing is nuclear translocation as well as its protein levels. Moreover, in assays with the inhibitor genistein, this transcription factor was found necessary for Epo-induced hepcidin suppression. Our findings support the involvement of the PI3K/AKT/mTOR pathway in the regulation of Fe levels by Epo, and highlight the contrasting roles of the C/EBP-α and HIF-1α transcription factors as downstream effectors of the cytokine in this process.
Collapse
Affiliation(s)
- Romina Eugenia Maltaneri
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina.
| | - María Eugenia Chamorro
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| | - Silvana Estela Gionco
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| | - Alcira Beatriz Nesse
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| | - Daniela Cecilia Vittori
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| |
Collapse
|
2
|
Dehghan Z, Mirmotalebisohi SA, Mozafar M, Sameni M, Saberi F, Derakhshanfar A, Moaedi J, Zohrevand H, Zali H. Deciphering the similarities and disparities of molecular mechanisms behind respiratory epithelium response to HCoV-229E and SARS-CoV-2 and drug repurposing, a systems biology approach. Daru 2024; 32:215-235. [PMID: 38652363 PMCID: PMC11087451 DOI: 10.1007/s40199-024-00507-0] [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: 09/17/2022] [Accepted: 02/08/2024] [Indexed: 04/25/2024] Open
Abstract
PURPOSE Identifying the molecular mechanisms behind SARS-CoV-2 disparities and similarities will help find new treatments. The present study determines networks' shared and non-shared (specific) crucial elements in response to HCoV-229E and SARS-CoV-2 viruses to recommend candidate medications. METHODS We retrieved the omics data on respiratory cells infected with HCoV-229E and SARS-CoV-2, constructed PPIN and GRN, and detected clusters and motifs. Using a drug-gene interaction network, we determined the similarities and disparities of mechanisms behind their host response and drug-repurposed. RESULTS CXCL1, KLHL21, SMAD3, HIF1A, and STAT1 were the shared DEGs between both viruses' protein-protein interaction network (PPIN) and gene regulatory network (GRN). The NPM1 was a specific critical node for HCoV-229E and was a Hub-Bottleneck shared between PPI and GRN in HCoV-229E. The HLA-F, ADCY5, TRIM14, RPF1, and FGA were the seed proteins in subnetworks of the SARS-CoV-2 PPI network, and HSPA1A and RPL26 proteins were the seed in subnetworks of the PPI network of HCOV-229E. TRIM14, STAT2, and HLA-F played the same role for SARS-CoV-2. Top enriched KEGG pathways included cell cycle and proteasome in HCoV-229E and RIG-I-like receptor, Chemokine, Cytokine-cytokine, NOD-like receptor, and TNF signaling pathways in SARS-CoV-2. We suggest some candidate medications for COVID-19 patient lungs, including Noscapine, Isoetharine mesylate, Cycloserine, Ethamsylate, Cetylpyridinium, Tretinoin, Ixazomib, Vorinostat, Venetoclax, Vorinostat, Ixazomib, Venetoclax, and epoetin alfa for further in-vitro and in-vivo investigations. CONCLUSION We suggested CXCL1, KLHL21, SMAD3, HIF1A, and STAT1, ADCY5, TRIM14, RPF1, and FGA, STAT2, and HLA-F as critical genes and Cetylpyridinium, Cycloserine, Noscapine, Ethamsylate, Epoetin alfa, Isoetharine mesylate, Ribavirin, and Tretinoin drugs to study further their importance in treating COVID-19 lung complications.
Collapse
Affiliation(s)
- Zeinab Dehghan
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Amir Mirmotalebisohi
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Mozafar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Sameni
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Saberi
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Derakhshanfar
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center of Comparative and Experimental Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Javad Moaedi
- Center of Comparative and Experimental Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Zohrevand
- Student Research Committee, Department of Biomedical Engineering and Medical Physics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biomedical Engineering and Medical Physics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Berna-Erro A, Granados MP, Rosado JA, Redondo PC. Thrombotic Alterations under Perinatal Hypoxic Conditions: HIF and Other Hypoxic Markers. Int J Mol Sci 2023; 24:14541. [PMID: 37833987 PMCID: PMC10572648 DOI: 10.3390/ijms241914541] [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: 07/28/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 10/15/2023] Open
Abstract
Hypoxia is considered to be a stressful physiological condition, which may occur during labor and the later stages of pregnancy as a result of, among other reasons, an aged placenta. Therefore, when gestation or labor is prolonged, low oxygen supply to the tissues may last for minutes, and newborns may present breathing problems and may require resuscitation maneuvers. As a result, poor oxygen supply to tissues and to circulating cells may last for longer periods of time, leading to life-threatening conditions. In contrast to the well-known platelet activation that occurs after reperfusion of the tissues due to an ischemia/reperfusion episode, platelet alterations in response to reduced oxygen exposition following labor have been less frequently investigated. Newborns overcome temporal hypoxic conditions by changing their organ functions or by adaptation of the intracellular molecular pathways. In the present review, we aim to analyze the main platelet modifications that appear at the protein level during hypoxia in order to highlight new platelet markers linked to complications arising from temporal hypoxic conditions during labor. Thus, we demonstrate that hypoxia modifies the expression and activity of hypoxic-response proteins (HRPs), including hypoxia-induced factor (HIF-1), endoplasmic reticulum oxidase 1 (Ero1), and carbonic anhydrase (CIX). Finally, we provide updates on research related to the regulation of platelet function due to HRP activation, as well as the role of HRPs in intracellular Ca2+ homeostasis.
Collapse
Affiliation(s)
- Alejandro Berna-Erro
- Department of Physiology (Phycell), University of Extremadura, Avd de la Universidad s/n, 10003 Caceres, Spain; (A.B.-E.); (P.C.R.)
| | | | - Juan Antonio Rosado
- Department of Physiology (Phycell), University of Extremadura, Avd de la Universidad s/n, 10003 Caceres, Spain; (A.B.-E.); (P.C.R.)
| | - Pedro Cosme Redondo
- Department of Physiology (Phycell), University of Extremadura, Avd de la Universidad s/n, 10003 Caceres, Spain; (A.B.-E.); (P.C.R.)
| |
Collapse
|
4
|
Zhou J, Lin Y, Kang X, Liu Z, Zou J, Xu F. Hypoxia-mediated promotion of glucose metabolism in non-small cell lung cancer correlates with activation of the EZH2/FBXL7/PFKFB4 axis. Cell Death Dis 2023; 14:326. [PMID: 37179372 PMCID: PMC10182982 DOI: 10.1038/s41419-023-05795-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 05/15/2023]
Abstract
F-box/LRR-repeat protein 7 (FBXL7) was predicted as a differentially expressed E3 ubiquitin ligase in non-small cell lung cancer (NSCLC), which has been suggested to influence cancer growth and metastasis. In this study, we aimed to decipher the function of FBXL7 in NSCLC and delineate the upstream and downstream mechanisms. FBXL7 expression was verified in NSCLC cell lines and GEPIA-based tissue samples, after which the upstream transcription factor of FBXL7 was bioinformatically identified. The substrate PFKFB4 of the FBXL7 was screened out by tandem affinity purification coupled with mass-spectrometry (TAP/MS). FBXL7 was downregulated in NSCLC cell lines and tissue samples. FBXL7 ubiquitinated and degraded PFKFB4, thus suppressing glucose metabolism and malignant phenotypes of NSCLC cells. Hypoxia-induced HIF-1α upregulation elevated EZH2 and inhibited FBXL7 transcription and reduced its expression, thus promoting PFKFB4 protein stability. By this mechanism, glucose metabolism and the malignant phenotype were enhanced. In addition, knockdown of EZH2 impeded tumor growth through the FBXL7/PFKFB4 axis. In conclusion, our work reveals that the EZH2/FBXL7/PFKFB4 axis plays a regulatory role in glucose metabolism and tumor growth of NSCLC, which is expected to be potential biomarkers for NSCLC.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yang Lin
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiuhua Kang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Zhicheng Liu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Juntao Zou
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Fei Xu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
5
|
Waseem A, Rashid S, Rashid K, Khan MA, Khan R, Haque R, Seth P, Raza SS. Insight into the transcription factors regulating Ischemic Stroke and Glioma in Response to Shared Stimuli. Semin Cancer Biol 2023; 92:102-127. [PMID: 37054904 DOI: 10.1016/j.semcancer.2023.04.006] [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: 11/23/2022] [Revised: 03/28/2023] [Accepted: 04/09/2023] [Indexed: 04/15/2023]
Abstract
Cerebral ischemic stroke and glioma are the two leading causes of patient mortality globally. Despite physiological variations, 1 in 10 people who have an ischemic stroke go on to develop brain cancer, most notably gliomas. In addition, glioma treatments have also been shown to increase the risk of ischemic strokes. Stroke occurs more frequently in cancer patients than in the general population, according to traditional literature. Unbelievably, these events share multiple pathways, but the precise mechanism underlying their co-occurrence remains unknown. Transcription factors (TFs), the main components of gene expression programmes, finally determine the fate of cells and homeostasis. Both ischemic stroke and glioma exhibit aberrant expression of a large number of TFs, which are strongly linked to the pathophysiology and progression of both diseases. The precise genomic binding locations of TFs and how TF binding ultimately relates to transcriptional regulation remain elusive despite a strong interest in understanding how TFs regulate gene expression in both stroke and glioma. As a result, the importance of continuing efforts to understand TF-mediated gene regulation is highlighted in this review, along with some of the primary shared events in stroke and glioma.
Collapse
Affiliation(s)
- Arshi Waseem
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow-226003, India
| | - Sumaiya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Khalid Rashid
- Department of Cancer Biology, Vontz Center for Molecular Studies, Cincinnati, OH 45267-0521
| | | | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City,Mohali, Punjab 140306, India
| | - Rizwanul Haque
- Department of Biotechnology, Central University of South Bihar, Gaya -824236, India
| | - Pankaj Seth
- Molecular and Cellular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Haryana-122052, India
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow-226003, India; Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College Hospital, Era University, Sarfarazganj, Lucknow-226003, India
| |
Collapse
|
6
|
Lai XX, Zhang CP, Wu YX, Yang Y, Zhang MQ, Qin WJ, Wang RX, Shu H. Comparative transcriptome analysis reveals physiological responses in liver tissues of Epinephelus coioides under acute hypoxia stress. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2022; 43:101005. [PMID: 35653833 DOI: 10.1016/j.cbd.2022.101005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
Hypoxia is a common stressor for aquatic animals, including Epinephelus coioides, with a considerable impact on sustainable aquaculture. E. coioides is a widely consumed fish in China owing to its high nutritious value and taste. However, water hypoxia caused by high density culture process has become a great threat to E. coioides culture, and its response to hypoxia stress has not been discussed before. Therefore, the aim of this study was to examine the response of E. coioides to acute hypoxia using transcriptomic techniques. To this end, RNA sequencing was performed on the liver tissues of fish exposed to normoxic and hypoxic conditions for 1 h. The results presented 503 differentially expressed genes (DEGs) in the liver tissue of fish exposed to hypoxic condition compared with those in the normoxic group. Enrichment analysis using the Gene Ontology database showed that the DEGs were mainly enriched for functions related to cell apoptosis signaling pathways, insulin resistance, antioxidant enzymes, and glycolysis/gluconeogenesis signaling pathways. KEGG enrichment analysis showed that HIF-1, PI3K-AKT, IL-17, NF-kappa B, and MAPK signaling pathways were significantly enriched by the DEGs. The DEGs were mainly involved in immune response, inflammatory response, cell apoptosis regulation, energy metabolism, and substance metabolism. Additionally, the hypoxia response in E. coioides was mainly regulated via the PI3K-AKT-HIF-1 signaling axis. Overall, the findings of this study contribute to the understanding of hypoxia stress response in E. coioides, and provides target genes for breeding hypoxia-tolerant Epinephelus spp.
Collapse
Affiliation(s)
- Xing-Xing Lai
- School of Life Sciences, Guangzhou University, Guangzhou 51006, China.
| | - Cui-Ping Zhang
- School of Life Sciences, Guangzhou University, Guangzhou 51006, China
| | - Yu-Xin Wu
- School of Life Sciences, Guangzhou University, Guangzhou 51006, China
| | - Yang Yang
- School of Life Sciences, Sun Yat-sen University, Guangzhou 51006, China
| | - Ming-Qing Zhang
- School of Life Sciences, Guangzhou University, Guangzhou 51006, China
| | - Wei-Jian Qin
- School of Life Sciences, Guangzhou University, Guangzhou 51006, China
| | - Rui-Xuan Wang
- School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou 521041, China.
| | - Hu Shu
- School of Life Sciences, Guangzhou University, Guangzhou 51006, China.
| |
Collapse
|
7
|
Xia B, Sui Q, Du Y, Wang L, Jing J, Zhu L, Zhao X, Sun X, Booth AM, Chen B, Qu K, Xing B. Secondary PVC microplastics are more toxic than primary PVC microplastics to Oryzias melastigma embryos. JOURNAL OF HAZARDOUS MATERIALS 2022; 424:127421. [PMID: 34653869 DOI: 10.1016/j.jhazmat.2021.127421] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 06/13/2023]
Abstract
Irregular-shaped and partially degraded secondary microplastics (SMP) account for the majority of MPs in marine environments, yet little is known about their effects on marine organisms. In this study, we investigated the embryotoxicity of polyvinyl chloride SMP and primary microplastics (PMP) to the marine medaka Oryzias melastigma. This study aimed to determine the physical impacts of MPs and, for the first time, elucidate the underlying mechanisms of physical toxicity. SMP shortened hatching time and induced higher teratogenic effects on larvae relative to PMP, indicating a higher toxicity from SMP. Physical damage from SMP to the chorion surface appears to be the main toxicity mechanism, caused by their irregular shape and reduced aggregation relative to PMP. In contrast, real-time changes in oxygen demonstrated that hypoxia caused by greater PMP adsorption to the chorion surface contributes to the toxicological responses of this material relative to SMP. Modulation of genes involved in hypoxia-response, cardiac development and hatching confirmed the toxicity mechanisms of PMP and SMP. The chemical contribution to observed toxicity was negligible, confirming impacts derived from physical toxicity. Our findings highlight the negative effects of environmentally relevant SMP on the marine ecosystems.
Collapse
Affiliation(s)
- Bin Xia
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266000, China.
| | - Qi Sui
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Yushan Du
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; College of Marine Sciences, Shanghai Ocean University, Shanghai 201306, China
| | - Liang Wang
- SINTEF Energy Research, Trondheim, 7034, Norway
| | - Jing Jing
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Lin Zhu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266000, China
| | - Xinguo Zhao
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266000, China
| | - Xuemei Sun
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266000, China
| | - Andy M Booth
- SINTEF Ocean, Department of Climate and Environment, Trondheim, 7465, Norway.
| | - Bijuan Chen
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266000, China
| | - Keming Qu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Baoshan Xing
- Stockbridge School of Agriculture, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
8
|
Lan YL, Zhu Y, Chen G, Zhang J. The Promoting Effect of Traumatic Brain Injury on the Incidence and Progression of Glioma: A Review of Clinical and Experimental Research. J Inflamm Res 2021; 14:3707-3720. [PMID: 34377008 PMCID: PMC8350857 DOI: 10.2147/jir.s325678] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 07/23/2021] [Indexed: 12/16/2022] Open
Abstract
The role of traumatic brain injury in the development of glioma is highly controversial since first presented. This is not unexpected because traumatic brain injuries are overwhelmingly more common than glioma. However, the causes of post-traumatic glioma have been long discussed and still warrant further research. In this review, we have presented an overview of previous cohort studies and case–control studies. We have summarized the roles of microglial cells, macrophages, astrocytes, and stem cells in post-traumatic glioma formation and development, and reviewed various carcinogenic factors involved during traumatic brain injury, especially those reported in experimental studies indicating a relationship with glioma progression. Besides, traumatic brain injury and glioma share several common pathways, including inflammation and oxidative stress; however, the exact mechanism underlying this co-occurrence is yet to be discovered. In this review, we have summarized current epidemiological studies, clinical reports, pathophysiological research, as well as investigations evaluating the probable causes of co-occurrence and treatment possibilities. More efforts should be directed toward elucidating the relationship between traumatic brain injury and glioma, which could likely lead to promising pharmacological interventions towards designing therapeutic strategies.
Collapse
Affiliation(s)
- Yu-Long Lan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, People's Republic of China
| | - Yongjian Zhu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
9
|
Chen X, Sun W, Zhong P, Wu D. Colony-Stimulating Factors on Mobilizing CD34 + Cells and Improving Neurological Functions in Patients With Stroke: A Meta-Analysis and a Systematic Review. Front Pharmacol 2021; 12:704509. [PMID: 34366857 PMCID: PMC8339259 DOI: 10.3389/fphar.2021.704509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/07/2021] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose: CSF therapy is considered a promising therapeutic approach for stroke. We performed a meta-analysis to explore the safety and efficacy of CSF in published clinical stroke studies. Methods: We searched articles online and manually. Two reviewers selected studies independently, selecting data based on study quality, characteristics of intervention (administration time, observation time, type, dose, and injection approach of CSF), and the baseline characteristics of patients (age, sex, hypertension, diabetes, smoker, and lipids) were extracted. Main prognosis outcomes were measured as all-cause death in severe adverse events (SAE) and recurrent stroke in SAE. Secondary outcomes were measured as CD34+ cell counts in periphery blood at day 5, National Institutes of Health Stroke Scale (NIHSS), and Barthel index (BI), Side effects of CSF were taken as the indicator of safety. STATA13 software was used to perform the meta-analysis.Keywords: Stroke, Colony-stimulating factor, Meta-analysis, therapy, Neurological Diseases Results: This meta-analysis involved 485 patients from eight studies. Among them, 475 patients from seven studies were gauged SAE (all-cause death), 393 patients from six studies were checked SAE (recurrent stroke); 137 patients from three studies underwent CD34+ measurement, 389 patients from six studies were tested NIHSS and 307 patients from five studies accessed BI. Compared with the control group, both all-causes death (RR= 1.73, 95%CI= (0.61, 4.92), P=0.735, I2=0.0%) and recurrent stroke (RR= 0.43, 95%CI= (0.14, 1.32), P=0.214, I2=33.1%) present no statistical differences, indicating that the application of CSF does not statistically alter the prognosis of patients with stroke. The application of CSF effectively enhanced CD34+ cell counts in periphery blood at day 5 (SMD= 1.23, 95%CI= (0.54, 1.92), P=0.04, I2=69.0%) but did not statistically impact NIHSS (SMD= -0.40, 95%CI= (-0.93, 0.13), P ≤ 0.001, I2=79.7%) or BI (SMD= 0.04, 95%CI= (-0.38, 0.46), P=0.068, I2=54.3%). Conclusion: Our study consolidates the security of CSF administration for its exerting no effect on detrimental outcomes. It has proven to be effective in elevating CD34+ cell counts in periphery blood at day 5, indicating CSF may participate in stroke recovery, but its efficacy in stroke recovery remains detected.
Collapse
Affiliation(s)
- Xiuqi Chen
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Wenbo Sun
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Ping Zhong
- Department of Neurology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Danhong Wu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Wang B, Zhang C, Chu D, Ma X, Yu T, Liu X, Hu C. Astragaloside IV improves angiogenesis under hypoxic conditions by enhancing hypoxia‑inducible factor‑1α SUMOylation. Mol Med Rep 2021; 23:244. [PMID: 33537820 PMCID: PMC7893755 DOI: 10.3892/mmr.2021.11883] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 10/08/2020] [Indexed: 12/18/2022] Open
Abstract
Improving angiogenic capacity under hypoxic conditions is essential for improving the survival of skin grafts, as they often lack the necessary blood supply. The stable expression levels of hypoxia‑inducible factor‑1α (HIF‑1α) in the nucleus directly affect the downstream vascular endothelial growth factor (VEGF) signaling pathway and regulate angiogenesis in a hypoxic environment. Astragaloside IV (AS‑IV), an active component isolated from Astragalus membranaceus, has multiple biological effects including antioxidant and anti‑diabetic effects, and the ability to provide protection from cardiovascular damage. However, the mechanisms underlying these effects have not previously been elucidated. The present study investigated whether AS‑IV promotes angiogenesis via affecting the balance between ubiquitination and small ubiquitin‑related modifier (SUMO) modification of HIF‑1α. The results demonstrated that persistent hypoxia induces changes in expression levels of HIF‑1α protein and significantly increases the proportion of dysplastic blood vessels. Further western blotting experiments showed that rapid attenuation and delayed compensation of SUMO1 activity is one of the reasons for the initial increase then decrease in HIF‑1α levels. SUMO1 overexpression stabilized the presence of HIF‑1α in the nucleus and decreased the extent of abnormal blood vessel morphology observed following hypoxia. AS‑IV induces vascular endothelial cells to continuously produce SUMO1, stabilizes the HIF‑1α/VEGF pathway and improves angiogenesis in hypoxic conditions. In summary, the present study confirmed that AS‑IV stimulates vascular endothelial cells to continuously resupply SUMO1, stabilizes the presence of HIF‑1α protein and improves angiogenesis in adverse hypoxic conditions, which may improve the success rate of flap graft surgery following trauma or burn.
Collapse
Affiliation(s)
- Baoshen Wang
- Department of Cardiovascular Surgery, The No. 1 Central Hospital of Baoding City, Baoding, Hebei 071000, P.R. China
| | - Chunyan Zhang
- Department of Pharmacy, Tianjin Binhai New Area Hospital of Traditional Chinese Medicine, Tianjin 300450, P.R. China
| | - Dongmei Chu
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin 300450, P.R. China
| | - Xiaofang Ma
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin 300450, P.R. China
| | - Tian Yu
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin 300450, P.R. China
| | - Xiaozhi Liu
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin 300450, P.R. China
| | - Changqing Hu
- The Fifth Orthopaedics Department, The No. 1 Central Hospital of Baoding City, Baoding, Hebei 071000, P.R. China
| |
Collapse
|
11
|
Auzmendi J, Puchulu MB, Rodríguez JCG, Balaszczuk AM, Lazarowski A, Merelli A. EPO and EPO-Receptor System as Potential Actionable Mechanism for the Protection of Brain and Heart in Refractory Epilepsy and SUDEP. Curr Pharm Des 2020; 26:1356-1364. [PMID: 32072891 DOI: 10.2174/1381612826666200219095548] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/31/2019] [Indexed: 12/26/2022]
Abstract
The most important activity of erythropoietin (EPO) is the regulation of erythrocyte production by activation of the erythropoietin receptor (EPO-R), which triggers the activation of anti-apoptotic and proliferative responses of erythroid progenitor cells. Additionally, to erythropoietic EPO activity, an antiapoptotic effect has been described in a wide spectrum of tissues. EPO low levels are found in the central nervous system (CNS), while EPO-R is expressed in most CNS cell types. In spite of EPO-R high levels expressed during the hypoxicischemic brain, insufficient production of endogenous cerebral EPO could be the cause of determined circuit alterations that lead to the loss of specific neuronal populations. In the heart, high EPO-R expression in cardiac progenitor cells appears to contribute to myocardial regeneration under EPO stimulation. Several lines of evidence have linked EPO to an antiapoptotic role in CNS and in heart tissue. In this review, an antiapoptotic role of EPO/EPO-R system in both brain and heart under hypoxic conditions, such as epilepsy and sudden death (SUDEP) has been resumed. Additionally, their protective effects could be a new field of research and a novel therapeutic strategy for the early treatment of these conditions and avoid SUDEP.
Collapse
Affiliation(s)
- Jerónimo Auzmendi
- Universidad de Buenos Aire (UBA), Facultad de Farmacia y Bioquimica (FFyB), Instituto de Fisiopatologia y Bioquimica Clínica (INFIBIOC), Junín 956, Ciudad Autonoma de Buenos Aires (CABA), Buenos Aires, Argentina
| | - María B Puchulu
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica, Departamento de Ciencias Biologicas, Catedra de Fisiologia, Instituto de Quimica y Metabolismo del Farmaco, CONICET, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina
| | - Julio C G Rodríguez
- CENPALAB, Centro Nacional para la Producción de Animales de Laboratorio, La Habana, Cuba
| | - Ana M Balaszczuk
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica, Departamento de Ciencias Biologicas, Catedra de Fisiologia, Instituto de Quimica y Metabolismo del Farmaco, CONICET, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Lazarowski
- Universidad de Buenos Aire (UBA), Facultad de Farmacia y Bioquimica (FFyB), Instituto de Fisiopatologia y Bioquimica Clínica (INFIBIOC), Junín 956, Ciudad Autonoma de Buenos Aires (CABA), Buenos Aires, Argentina
| | - Amalia Merelli
- Universidad de Buenos Aire (UBA), Facultad de Farmacia y Bioquimica (FFyB), Instituto de Fisiopatologia y Bioquimica Clínica (INFIBIOC), Junín 956, Ciudad Autonoma de Buenos Aires (CABA), Buenos Aires, Argentina
| |
Collapse
|
12
|
Gamdzyk M, Doycheva DM, Kang R, Tang H, Travis ZD, Tang J, Zhang JH. GW0742 activates miR-17-5p and inhibits TXNIP/NLRP3-mediated inflammation after hypoxic-ischaemic injury in rats and in PC12 cells. J Cell Mol Med 2020; 24:12318-12330. [PMID: 33034416 PMCID: PMC7686982 DOI: 10.1111/jcmm.15698] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/23/2020] [Accepted: 07/09/2020] [Indexed: 12/19/2022] Open
Abstract
This study aimed to investigate the effects of PPAR‐β/δ receptor agonist GW0742 on neuroinflammation in a rat model of hypoxia‐ischaemia (HI) and in PC12 cells in OGD model. HI was induced by ligating the common carotid artery and inducing hypoxia for 150 minutes. Immunofluorescence was used for quantification of microglia activation and for determining cellular localization of PPAR‐β/δ. Expression of proteins was measured by Western blot. Activation of miR‐17‐5p by GW0742 was assessed in PC12 cells by Dual‐Luciferase Reporter Gene Assay. The endogenous expression of TXNIP, NLRP3, cleaved caspase‐1 and IL‐1β was increased after HI. GW0742 treatment significantly reduced the number of activated pro‐inflammatory microglia in ipsilateral hemisphere after HI. Mechanistically, GW0742 significantly decreased the expression of TXNIP, NLRP3, IL‐6 and TNF‐α. Either PPAR‐β/δ antagonist GSK3787, miR‐17‐5p inhibitor, or TXNIP CRISPR activation abolished the anti‐inflammatory effects of GW0742. Activation of PPAR‐β/δ by GW0742 activated miR‐17‐5p expression in PC12 cells and increased cell viability after OGD, which was accompanied by decreased expression of TXNIP and reduced secretion of IL‐1β and TNF‐α. In conclusion, GW0742 may be a promising neurotherapeutic for the management of HI patients.
Collapse
Affiliation(s)
- Marcin Gamdzyk
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Desislava Met Doycheva
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Ruiqing Kang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Hong Tang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Zackary D Travis
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Anesthesiology, Neurosurgery and Neurology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
13
|
Sun X, Liu XZ, Wang J, Tao HR, Zhu T, Jin WJ, Shen KP. Changes in neurological and pathological outcomes in a modified rat spinal cord injury model with closed canal. Neural Regen Res 2020; 15:697-704. [PMID: 31638094 PMCID: PMC6975156 DOI: 10.4103/1673-5374.266919] [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] [Indexed: 12/17/2022] Open
Abstract
Most animal spinal cord injury models involve a laminectomy, such as the weight drop model or the transection model. However, in clinical practice, many patients undergo spinal cord injury while maintaining a relatively complete spinal canal. Thus, open spinal cord injury models often do not simulate real injuries, and few previous studies have investigated whether having a closed spinal canal after a primary spinal cord injury may influence secondary processes. Therefore, we aimed to assess the differences in neurological dysfunction and pathological changes between rat spinal cord injury models with closed and open spinal canals. Sprague-Dawley rats were randomly divided into three groups. In the sham group, the tunnel was expanded only, without inserting a screw into the spinal canal. In the spinal cord injury with open canal group, a screw was inserted into the spinal canal to cause spinal cord injury for 5 minutes, and then the screw was pulled out, leaving a hole in the vertebral plate. In the spinal cord injury with closed canal group, after inserting a screw into the spinal canal for 5 minutes, the screw was pulled out by approximately 1.5 mm and the flat end of the screw remained in the hole in the vertebral plate so that the spinal canal remained closed; this group was the modified model, which used a screw both to compress the spinal cord and to seal the spinal canal. At 7 days post-operation, the Basso-Beattie-Bresnahan scale was used to measure changes in neurological outcomes. Hematoxylin-eosin staining was used to assess histopathology. To evaluate the degree of local secondary hypoxia, immunohistochemical staining and western blot assays were applied to detect the expression of hypoxia-inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF). Compared with the spinal cord injury with open canal group, in the closed canal group the Basso-Beattie-Bresnahan scores were lower, cell morphology was more irregular, the percentage of morphologically normal neurons was lower, the percentages of HIF-1α- and VEGF-immunoreactive cells were higher, and HIF-1α and VEGF protein expression was also higher. In conclusion, we successfully established a rat spinal cord injury model with closed canal. This model could result in more serious neurological dysfunction and histopathological changes than in open canal models. All experimental procedures were approved by the Institutional Animal Care Committee of Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, China (approval No. HKDL201810) on January 30, 2018.
Collapse
Affiliation(s)
- Xin Sun
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xing-Zhen Liu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Wang
- Department of Pathology, Shanghai Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai-Rong Tao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tong Zhu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Jie Jin
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kang-Ping Shen
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Doycheva D, Xu N, Kaur H, Malaguit J, McBride DW, Tang J, Zhang JH. Adenoviral TMBIM6 vector attenuates ER-stress-induced apoptosis in a neonatal hypoxic-ischemic rat model. Dis Model Mech 2019; 12:dmm040352. [PMID: 31636086 PMCID: PMC6898997 DOI: 10.1242/dmm.040352] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/14/2019] [Indexed: 12/31/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is a major pathology encountered after hypoxic-ischemic (HI) injury. Accumulation of unfolded proteins triggers the unfolded protein response (UPR), resulting in the activation of pro-apoptotic cascades that lead to cell death. Here, we identified Bax inhibitor 1 (BI-1), an evolutionarily conserved protein encoded by the transmembrane BAX inhibitor motif-containing 6 (TMBIM6) gene, as a novel modulator of ER-stress-induced apoptosis after HI brain injury in a neonatal rat pup. The main objective of our study was to overexpress BI-1, via viral-mediated gene delivery of human adenoviral-TMBIM6 (Ad-TMBIM6) vector, to investigate its anti-apoptotic effects as well as to elucidate its signaling pathways in an in vivo neonatal HI rat model and in vitro oxygen-glucose deprivation (OGD) model. Ten-day-old unsexed Sprague Dawley rat pups underwent right common carotid artery ligation followed by 1.5 h of hypoxia. Rat pups injected with Ad-TMBIM6 vector, 48 h pre-HI, showed a reduction in relative infarcted area size, attenuated neuronal degeneration and improved long-term neurological outcomes. Furthermore, silencing of BI-1 or further activating the IRE1α branch of the UPR, using a CRISPR activation plasmid, was shown to reverse the protective effects of BI-1. Based on our in vivo and in vitro data, the protective effects of BI-1 are mediated via inhibition of IRE1α signaling and in part via inhibition of the second stress sensor receptor, PERK. Overall, this study showed a novel role for BI-1 and ER stress in the pathophysiology of HI and could provide a basis for BI-1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Desislava Doycheva
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Ningbo Xu
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
- Department of Interventional Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Harpreet Kaur
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Jay Malaguit
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Devin William McBride
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jiping Tang
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - John H Zhang
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
- Departments of Anesthesiology, Neurosurgery and Neurology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| |
Collapse
|
15
|
Ghosh MK, Chakraborty D, Sarkar S, Bhowmik A, Basu M. The interrelationship between cerebral ischemic stroke and glioma: a comprehensive study of recent reports. Signal Transduct Target Ther 2019; 4:42. [PMID: 31637020 PMCID: PMC6799849 DOI: 10.1038/s41392-019-0075-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/26/2019] [Accepted: 08/29/2019] [Indexed: 12/16/2022] Open
Abstract
Glioma and cerebral ischemic stroke are two major events that lead to patient death worldwide. Although these conditions have different physiological incidences, ~10% of ischemic stroke patients develop cerebral cancer, especially glioma, in the postischemic stages. Additionally, the high proliferation, venous thrombosis and hypercoagulability of the glioma mass increase the significant risk of thromboembolism, including ischemic stroke. Surprisingly, these events share several common pathways, viz. hypoxia, cerebral inflammation, angiogenesis, etc., but the proper mechanism behind this co-occurrence has yet to be discovered. The hypercoagulability and presence of the D-dimer level in stroke are different in cancer patients than in the noncancerous population. Other factors such as atherosclerosis and coagulopathy involved in the pathogenesis of stroke are partially responsible for cancer, and the reverse is also partially true. Based on clinical and neurosurgical experience, the neuronal structures and functions in the brain and spine are observed to change after a progressive attack of ischemia that leads to hypoxia and atrophy. The major population of cancer cells cannot survive in an adverse ischemic environment that excludes cancer stem cells (CSCs). Cancer cells in stroke patients have already metastasized, but early-stage cancer patients also suffer stroke for multiple reasons. Therefore, stroke is an early manifestation of cancer. Stroke and cancer share many factors that result in an increased risk of stroke in cancer patients, and vice-versa. The intricate mechanisms for stroke with and without cancer are different. This review summarizes the current clinical reports, pathophysiology, probable causes of co-occurrence, prognoses, and treatment possibilities.
Collapse
Affiliation(s)
- Mrinal K. Ghosh
- Signal Transduction in Cancer and Stem Cells Laboratory, Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), 4 Raja S.C. Mullick Road, Kolkata 700032 and CN-06, Sector-V, Salt Lake, Kolkata, 700091 India
| | - Dipankar Chakraborty
- Signal Transduction in Cancer and Stem Cells Laboratory, Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), 4 Raja S.C. Mullick Road, Kolkata 700032 and CN-06, Sector-V, Salt Lake, Kolkata, 700091 India
| | - Sibani Sarkar
- Signal Transduction in Cancer and Stem Cells Laboratory, Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), 4 Raja S.C. Mullick Road, Kolkata 700032 and CN-06, Sector-V, Salt Lake, Kolkata, 700091 India
| | - Arijit Bhowmik
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37 S. P. Mukherjee Road, Kolkata, 700 026 India
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, Dakshin Barasat, South 24, Paraganas, 743372 India
| |
Collapse
|
16
|
Doycheva D, Xu N, Tang J, Zhang J. Viral-mediated gene delivery of TMBIM6 protects the neonatal brain via disruption of NPR-CYP complex coupled with upregulation of Nrf-2 post-HI. J Neuroinflammation 2019; 16:174. [PMID: 31472686 PMCID: PMC6717394 DOI: 10.1186/s12974-019-1559-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 08/15/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Oxidative stress, inflammation, and endoplasmic reticulum (ER) stress play a major role in the pathogenesis of neonatal hypoxic-ischemic (HI) injury. ER stress results in the accumulation of unfolded proteins that trigger the NADPH-P450 reductase (NPR) and the microsomal monooxygenase system which is composed of cytochrome P450 members (CYP) generating reactive oxygen species (ROS) as well as the release of inflammatory cytokines. We explored the role of Bax Inhibitor-1 (BI-1) protein, encoded by the Transmembrane Bax inhibitor Motif Containing 6 (TMBIM6) gene, in protection from ER stress after HI brain injury. BI-1 may attenuate ER stress-induced ROS production and release of inflammatory mediators via (1) disruption of the NPR-CYP complex and (2) upregulation of Nrf-2, a redox-sensitive transcription factor, thus promoting an increase in anti-oxidant enzymes to inhibit ROS production. The main objective of our study is to evaluate BI-1's inhibitory effects on ROS production and inflammation by overexpressing BI-1 in 10-day-old rat pups. METHODS Ten-day-old (P10) unsexed Sprague-Dawley rat pups underwent right common carotid artery ligation, followed by 1.5 h of hypoxia. To overexpress BI-1, rat pups were intracerebroventricularly (icv) injected at 48 h pre-HI with the human adenoviral vector-TMBIM6 (Ad-TMBIM6). BI-1 and Nrf-2 silencing were achieved by icv injection at 48 h pre-HI using siRNA to elucidate the potential mechanism. Percent infarcted area, immunofluorescent staining, DHE staining, western blot, and long-term neurobehavior assessments were performed. RESULTS Overexpression of BI-1 significantly reduced the percent infarcted area and improved long-term neurobehavioral outcomes. BI-1's mediated protection was observed to be via inhibition of P4502E1, a major contributor to ROS generation and upregulation of pNrf-2 and HO-1, which correlated with a decrease in ROS and inflammatory markers. This effect was reversed when BI-1 or Nrf-2 were inhibited. CONCLUSIONS Overexpression of BI-1 increased the production of antioxidant enzymes and attenuated inflammation by destabilizing the complex responsible for ROS production. BI-1's multimodal role in inhibiting P4502E1, together with upregulating Nrf-2, makes it a promising therapeutic target.
Collapse
Affiliation(s)
- Desislava Doycheva
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92354 USA
| | - Ningbo Xu
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92354 USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92354 USA
| | - John Zhang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92354 USA
- Departments of Anesthesiology, Neurosurgery and Neurology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Loma Linda, CA 92354 USA
| |
Collapse
|
17
|
Berlian G, Tandrasasmita OM, Tjandrawinata RR. Upregulation of endogenous erythropoietin expression by DLBS6747, a bioactive fraction of Ipomoea batatas L. leaves, via increasing HIF1α transcription factor in HEK293 kidney cells. JOURNAL OF ETHNOPHARMACOLOGY 2019; 235:190-198. [PMID: 30685435 DOI: 10.1016/j.jep.2019.01.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ipomoea batatas L., locally known as ubi jalar, is widely used in Indonesia and other countries as a folk remedy for various chronic diseases, including anemia-associated chronic kidney disease by increasing hematological parameters such as packed cell volume, white blood cells and platelet counts. AIM OF THE STUDY The aim of this study is to evaluate the effect of DLBS6747, a bioactive fraction of I. batatas L. leaves, on increasing EPO expression through the upregulation of HIF1α. MATERIALS AND METHODS Effect of DLBS6747 on EPO expression and its transcription factor, HIFs, was evaluated in normoxia and hypoxia conditions. Effect of DLBS6747 on several genes involved in EPO expression were evaluated in a time-course manner using conventional and real-time PCR, while the protein level were revealed using western blot and ELISA. The involvement of HIF1α was also confirmed by HIF1α siRNA. RESULTS Administration of DLBS6747 increased transcriptional activity of EPO through the regulation of its transcriptional factors, which include HIF1α, HIF2α and NFᴋB. The effect was found to be dependent on oxygen availability, wherein DLBS6747-increased EPO expression was found to be more significant in hypoxic condition. In normoxia and hypoxia, 40 μg/mL DLBS6747 increased HIF1α and HIF2α expressions at mRNA level, wherein the peak appeared in 12 h treatment (up to 7.9- and 8.6-folds, respectively). On the other hand, increased protein level was only found in hypoxia, where the highest HIF1α expression was observed at 6 h (7.5-folds increase) and started to decrease after the hours, while HIF2α was found to be increased time-dependently (up to 13.8-folds in 24 h). The mechanism of action of DLBS6747 as erythropoietin stimulating agent is more likely to affect the regulation of HIF1α, as confirmed by HIF1α siRNA which showed that DLBS6747 failed to increase EPO expression during co-incubation with HIF1α siRNA. DLBS6747 treatment also decreased NFᴋB time-dependently in normoxia, while no NFᴋB was detected in hypoxia, which revealed mimicking hypoxia activity of DLBS6747 to increase EPO expression. CONCLUSION These findings showed convincing evidences that DLBS6747 increases endogenous EPO production primarily via upregulation of its transcription factors, especially HIF1α, in human embryonic kidney HEK293 cells. This is the first molecular report that reveals the mechanism of action of natural-based erythropenia drug in different oxygen availability.
Collapse
Affiliation(s)
- Guntur Berlian
- Dexa Laboratories of Biomolecular Sciences (DLBS), Industri Selatan V Block PP No. 7, Jababeka Industrial Estate II, Cikarang 17550, West Java, Indonesia.
| | - Olivia Mayasari Tandrasasmita
- Dexa Laboratories of Biomolecular Sciences (DLBS), Industri Selatan V Block PP No. 7, Jababeka Industrial Estate II, Cikarang 17550, West Java, Indonesia.
| | - Raymond Rubianto Tjandrawinata
- Dexa Laboratories of Biomolecular Sciences (DLBS), Industri Selatan V Block PP No. 7, Jababeka Industrial Estate II, Cikarang 17550, West Java, Indonesia.
| |
Collapse
|
18
|
TRAF2 protects against cerebral ischemia-induced brain injury by suppressing necroptosis. Cell Death Dis 2019; 10:328. [PMID: 30988281 PMCID: PMC6465397 DOI: 10.1038/s41419-019-1558-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/21/2019] [Accepted: 03/25/2019] [Indexed: 12/12/2022]
Abstract
Necroptosis contributes to ischemia-induced brain injury. Tumor necrosis factor (TNF) receptor associated factor 2 (TRAF2) has been reported to suppress necroptotic cell death under several pathological conditions. In this study, we investigated the role of TRAF2 in experimental stroke using a mouse middle cerebral artery occlusion (MCAO) model and in vitro cellular models. TRAF2 expression in the ischemic brain was assessed with western blot and real-time RT-PCR. Gene knockdown of TRAF2 by lentivirus was utilized to investigate the role of TRAF2 in stroke outcomes. The expression of TRAF2 was significantly induced in the ischemic brain at 24 h after reperfusion, and neurons and microglia were two of the cellular sources of TRAF2 induction. Striatal knockdown of TRAF2 increased infarction size, cell death, microglial activation and the expression of pro-inflammatory markers at 24 h after reperfusion. TRAF2 expression and necroptosis were induced in mouse primary microglia treated with conditioned medium collected from neurons subject to oxygen and glucose deprivation (OGD) and in TNFα-treated mouse hippocampal neuronal HT-22 cells in the presence of the pan-caspase inhibitor Z-VAD. In addition, TRAF2 knockdown exacerbated microglial cell death and neuronal cell death under these conditions. Moreover, pre-treatment with a specific necroptosis inhibitor necrostatin-1 (nec-1) suppressed the cell death exacerbated by TRAF2 knockdown in the brain following MCAO, indicating that TRAF2 impacted ischemic brain damage through necroptosis mechanism. Taken together, our results demonstrate that TRAF2 is a novel regulator of cerebral ischemic injury.
Collapse
|
19
|
Erythropoietin protects neurons from apoptosis via activating PI3K/AKT and inhibiting Erk1/2 signaling pathway. 3 Biotech 2019; 9:131. [PMID: 30863710 DOI: 10.1007/s13205-019-1667-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 03/01/2019] [Indexed: 12/11/2022] Open
Abstract
The aim of this study was to explore the neuroprotective effect and the underlying mechanism of erythropoietin (EPO) on the cortical neuronal cells insulted with oxygen and glucose deprivation (OGD). Different concentrations of EPO were used to determine the anti-apoptosis effect of EPO. In addition, PI3K inhibitor LY294002 and ERK1/2 inhibitor U0126 were added to explore the underlying mechanism of EPO. Cell apoptosis rate was measured by flow cytometry. The protein expression of Bax, Bcl-2, cleaved caspase-3, AKT, p-AKT, Erk1/2 and p-Erk1/2 wasmeasured by Western blot. Our results showed that EPO alleviates OGD-induced cell apoptosis in a dose-dependent manner; the neuroprotective effect of EPO was further confirmed by the fact that EPO treatment reversed the protein expression of cleaved caspase-3, as well as the Bcl-2/Bax ratio as compared with the OGD treatment. In the mechanism part, our results demonstrated that OGD and EPO nearly had no influence on the protein expression of AKT and Erk1/2 but altered the phosphorylation of them. Specifically, OGD decreased the expression of p-AKT and increased the expression of p-Erk1/2; while, EPO treatment reversed the expression of p-AKT and p-Erk1/2 as compared with OGD treatment. Interestingly, LY294002 decreased the expression of p-AKT and attenuated the neuroprotective effect of EPO; while, U0126 decreased the expression of p-Erk1/2 and enhanced the neuroprotective effect of EPO. Our study demonstrated that EPO protects neurons against apoptosis induced by OGD, which is closely related with activation of PI3K/AKT and inactivation of Erk1/2 signaling pathway.
Collapse
|
20
|
O'Neill J, Jasionek G, Drummond SE, Brett O, Lucking EF, Abdulla MA, O'Halloran KD. Renal cortical oxygen tension is decreased following exposure to long-term but not short-term intermittent hypoxia in the rat. Am J Physiol Renal Physiol 2019; 316:F635-F645. [PMID: 30648908 DOI: 10.1152/ajprenal.00254.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chronic kidney disease (CKD) occurs in more than 50% of patients with obstructive sleep apnea (OSA). However, the impact of intermittent hypoxia (IH) on renal function and oxygen homeostasis is unclear. Male Sprague-Dawley rats were exposed to IH (270 s at 21% O2; 90 s hypoxia, 6.5% O2 at nadir) for 4 h [acute IH (AIH)] or to chronic IH (CIH) for 8 h/day for 2 wk. Animals were anesthetized and surgically prepared for the measurement of mean arterial pressure (MAP), and left renal excretory function, renal blood flow (RBF), and renal oxygen tension (Po2). AIH had no effect on MAP (123 ± 14 vs. 129 ± 14 mmHg, means ± SE, sham vs. IH). The CIH group was hypertensive (122 ± 9 vs. 144 ± 15 mmHg, P < 0.05). Glomerular filtration rate (GFR) (0.92 ± 0.27 vs. 1.33 ± 0.33 ml/min), RBF (3.8 ± 1.5 vs. 7.2 ± 2.4 ml/min), and transported sodium (TNa) (132 ± 39 vs. 201 ± 47 μmol/min) were increased in the AIH group (all P < 0.05). In the CIH group, GFR (1.25 ± 0.28 vs. 0.86 ± 0.28 ml/min, P < 0.05) and TNa (160 ± 39 vs. 120 ± 40 μmol/min, P < 0.05) were decreased, while RBF (4.13 ± 1.5 vs. 3.08 ± 1.5 ml/min) was not significantly different. Oxygen consumption (QO2) was increased in the AIH group (6.76 ± 2.60 vs. 13.60 ± 7.77 μmol/min, P < 0.05), but it was not significantly altered in the CIH group (3.97 ± 2.63 vs. 6.82 ± 3.29 μmol/min). Cortical Po2 was not significantly different in the AIH group (46 ± 4 vs. 46 ± 3 mmHg), but it was decreased in the CIH group (44 ± 5 mmHg vs. 38 ± 2 mmHg, P < 0.05). For AIH, renal oxygen homeostasis was preserved through a maintained balance between O2 supply (RBF) and consumption (GFR). For CIH, mismatched TNa and QO2 reflect inefficient O2 utilization and, thereby, sustained decrease in cortical Po2.
Collapse
Affiliation(s)
- Julie O'Neill
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| | - Greg Jasionek
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| | - Sarah E Drummond
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| | - Orla Brett
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| | - Eric F Lucking
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| | - Mohammed A Abdulla
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| |
Collapse
|
21
|
Sheikh AM, Yano S, Mitaki S, Haque MA, Yamaguchi S, Nagai A. A Mesenchymal stem cell line (B10) increases angiogenesis in a rat MCAO model. Exp Neurol 2018; 311:182-193. [PMID: 30291853 DOI: 10.1016/j.expneurol.2018.10.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 08/29/2018] [Accepted: 10/01/2018] [Indexed: 12/13/2022]
Abstract
A human mesenchymal stem cell line (B10) transplantation has been shown to improve ischemia-induced neurological deficits in animal stroke models. To understand the underlying mechanism, we have investigated the effects of B10 transplantation on cerebral angiogenesis in a rat middle cerebral artery occlusion (MCAO) model. B10 cells were transplanted intravenously 24 h after MCAO. Immunofluorescence staining results showed that compared to PBS-groups, vWF positive vessel and endoglin positive new vessels were increased in B10-transplanted MCAO groups in the lesion areas. The mRNA of angiogenesis factors including placental growth factor and hypoxia inducible factor (HIF)-1α were increased 3 days after MCAO in the core and IBZ areas of B10-transplanted group. Angiopoetin1 mRNA was increased only in the IBZ. Western blotting results showed that HIF-1α and vascular endothelial growth factor (VEGF) proteins were increased in B10-transplanted group. Both HIF-1α and VEGF were expressed in macrophage/microglia in the core area. In the IBZ, however, HIF-1α was expressed both in astrocytes and macrophage/microglia, while VEGF was expressed only in macrophage/microglia. Moreover, TGFβ protein levels were found to be increased in B10-transplanted group in the core and IBZ regions. Cell culture experiments using a human microglia cell line (HMO6) and B10 showed that IL-1β induced VEGF mRNA expression in both cell types. IL-1β was found to be highly expressed in B10 cells, and its co-culture with HMO6 further increased that in B10. Co-culture increased VEGF mRNA in both B10 and HMO6. In the rat brains, IL-1β was expressed in macrophage/microglia and transplanted-B10 cells in the core. IL-1β positive cell number was increased slightly, but significantly in B10-transplanted rats. To explore further, IL-1β expression was silenced in B10 cells by transfecting mRNA specific siRNA, and then transplanted in MCAO rats. Immunostaining result showed that endoglin positive area was decreased in IL-1β-silenced B10 transplanted groups compared to nonsilenced-B10 transplanted groups. Interestingly, vessel-like structure appeared as early as 3 days after MCAO in IL-1β-silenced B10-transplanted group. Thus our results demonstrated that B10 cells increased angiogenesis in MCAO rat model, through the regulation of HIF-1α and VEGF expression, where IL-1β might play a role.
Collapse
Affiliation(s)
- Abdullah Md Sheikh
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Shozo Yano
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Shingo Mitaki
- Department of Neurology, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Md Ahsanul Haque
- Department of Neurology, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Shuhei Yamaguchi
- Department of Neurology, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Atsushi Nagai
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan.
| |
Collapse
|
22
|
Shi X, Zhang G, Sun H, Bai Y, Liu Y, Zhang W, Xiao H. Effects of over-expression of HIF-1alpha in bone marrow-derived mesenchymal stem cells on traumatic brain injury. Eng Life Sci 2018; 18:401-407. [PMID: 32624920 DOI: 10.1002/elsc.201800015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 02/24/2018] [Accepted: 03/13/2018] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been shown to play therapeutic effect in traumatic brain injury (TBI). To augment the therapeutic effect, MSCs could be engineered to over-express genes that are beneficial for treatment. In the present study, we over-expressed hypoxia inducible factor (HIF)-1alpha in bone marrow derived MSCs (BM-MSCs) and sought to investigate whether HIF-1alpha could enhance the therapeutic effect of MSCs in a mouse model of TBI. Balb/c mice were subjected to controlled cortical impact injury and MSCs were transplanted intravenously at 6 h after injury. The lesion volume and brain water content were measured and the neurological function was assessed by modified neurologic severity score tests. Double-labeled immunofluorescence for BrdU and NeuU was performed to determine angiogenesis and neurogenesis. The expression of erythropoietin (EPO) and vascular endothelial growth factor (VEGF) was measured by quantitative RT-PCR and western blotting. After TBI, mice received BM-MSCs over-expressing HIF-1alpha showed significantly more functional recovery, reduced brain damage, increased angiogenesis and neurogenesis and increased expression of VEGF and EPO, compared with control mice or mice treated with non-transduced BM-MSCs. Over-expression of HIF-1alpha enhanced BM-MSCs induced improvement of neurological recovery after TBI, by stimulating angiogenesis and neurogenesis.
Collapse
Affiliation(s)
- Xiaodong Shi
- Qilu Hospital of Shandong University Jinan Shandong P. R. China.,Weifang Yidu Central Hospital Qingzhou Shandong P. R. China
| | - Guodong Zhang
- Weifang Yidu Central Hospital Qingzhou Shandong P. R. China
| | - Huijuan Sun
- Weifang Yidu Central Hospital Qingzhou Shandong P. R. China
| | - Yunan Bai
- Weifang Yidu Central Hospital Qingzhou Shandong P. R. China
| | - Yuguang Liu
- Qilu Hospital of Shandong University Jinan Shandong P. R. China
| | - Wei Zhang
- Weifang Yidu Central Hospital Qingzhou Shandong P. R. China
| | - Hang Xiao
- Weifang Yidu Central Hospital Qingzhou Shandong P. R. China
| |
Collapse
|
23
|
Neuroprotective Effects of Bioactive Compounds and MAPK Pathway Modulation in "Ischemia"-Stressed PC12 Pheochromocytoma Cells. Brain Sci 2018; 8:brainsci8020032. [PMID: 29419806 PMCID: PMC5836051 DOI: 10.3390/brainsci8020032] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/24/2018] [Accepted: 02/02/2018] [Indexed: 02/08/2023] Open
Abstract
This review surveys the efforts taken to investigate in vitro neuroprotective features of synthetic compounds and cell-released growth factors on PC12 clonal cell line temporarily deprived of oxygen and glucose followed by reoxygenation (OGD/R). These cells have been used previously to mimic some of the properties of in vivo brain ischemia-reperfusion-injury (IRI) and have been instrumental in identifying common mechanisms such as calcium overload, redox potential, lipid peroxidation and MAPKs modulation. In addition, they were useful for establishing the role of certain membrane penetrable cocktails of antioxidants as well as potential growth factors which may act in neuroprotection. Pharmacological mechanisms of neuroprotection addressing modulation of the MAPK cascade and increased redox potential by natural products, drugs and growth factors secreted by stem cells, in either undifferentiated or nerve growth factor-differentiated PC12 cells exposed to ischemic conditions are discussed for future prospects in neuroprotection studies.
Collapse
|
24
|
Interplay between mitochondrial metabolism and oxidative stress in ischemic stroke: An epigenetic connection. Mol Cell Neurosci 2017; 82:176-194. [DOI: 10.1016/j.mcn.2017.05.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/26/2017] [Accepted: 05/24/2017] [Indexed: 12/18/2022] Open
|
25
|
Li C, Zhang T, Yu K, Xie H, Bai Y, Zhang L, Wu Y, Wang N. Neuroprotective effect of electroacupuncture and upregulation of hypoxia-inducible factor-1α during acute ischaemic stroke in rats. Acupunct Med 2017; 35:360-365. [PMID: 28536255 DOI: 10.1136/acupmed-2016-011148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Acupuncture is a traditional method that has been widely used in various fields of medicine with therapeutic effect. However, evidence of effectiveness to support the application of electroacupuncture (EA) during the process of ischaemia is scarce. OBJECTIVES To investigate dynamic changes in hypoxia-inducible factor (HIF)-1α expression as well as its association with neurological status in rats subjected to acute ischaemic stroke and EA intervention. METHODS Forty adult male rats were randomly divided into three groups that received sham surgery (Control group, n=10) or underwent middle cerebral artery occlusion and EA (MCAO+EA group, n=15) or minimal acupuncture as a control treatment (MCAO+MA group, n=15). The rats in the MCAO+EA and MCAO+MA groups received EA or acupuncture without any electrical current, respectively, during 90 min of ischaemia. Rats in the Control group received the same surgical procedure but without MCAO. EA involved electrical stimulation of needles inserted into the quadriceps at 50 Hz frequency and 3 mA current intensity. Neurological status was evaluated on postoperative day 1, and cerebral infarction volume (IV) and HIF-1α expression 24 hours later. RESULTS Neurological scores were improved and cerebral IV was decreased in the MCAO+EA group compared to the MCAO+MA group (both p<0.05). Moreover, HIF-1α expression was higher in the MCAO+EA group versus the MCAO+MA group (p<0.05). CONCLUSIONS EA enhanced recovery of neurological function, decreased cerebral IV and increased HIF-1α expression in ischaemic rats. Further research is needed to determine whether EA is effective for stroke treatment through the stimulation of muscle contraction.
Collapse
Affiliation(s)
- Ce Li
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, WuLuMuQi Middle Road 12, Shanghai, China
| | - Tingting Zhang
- Acupuncture and Tuina College, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, China
| | - Kewei Yu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, WuLuMuQi Middle Road 12, Shanghai, China
| | - Hongyu Xie
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, WuLuMuQi Middle Road 12, Shanghai, China
| | - Yulong Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, WuLuMuQi Middle Road 12, Shanghai, China
| | - Li Zhang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, WuLuMuQi Middle Road 12, Shanghai, China
| | - Yi Wu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, WuLuMuQi Middle Road 12, Shanghai, China
| | - Nianhong Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, WuLuMuQi Middle Road 12, Shanghai, China
| |
Collapse
|
26
|
Endogenous regeneration: Engineering growth factors for stroke. Neurochem Int 2017; 107:57-65. [PMID: 28411103 DOI: 10.1016/j.neuint.2017.03.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/30/2017] [Accepted: 03/31/2017] [Indexed: 12/31/2022]
Abstract
Despite the efforts in developing therapeutics for stroke, recombinant tissue plasminogen activator (rtPA) remains the only FDA approved drug for ischemic stroke. Regenerative medicine targeting endogenous growth factors has drawn much interest in the clinical field as it provides potential restoration for the damaged brain tissue without being limited by a narrow therapeutic window. To date, most of the translational studies using regenerative medicines have encountered problems and failures. In this review, we discuss the effects of some trophic factors which include of erythropoietin (EPO), brain derived neurotrophic factor (BDNF), granulocyte-colony stimulating factor (G-CSF), vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), epidermal growth factor (EGF) and heparin binding epidermal growth factor (HB-EGF) in experimental ischemic stroke models and elaborate the lost in translation of the candidate growth factors from bench to bedside. Several new methodologies have been developed to overcome the caveats in translational studies. This review highlights the latest bioengineering approaches including the controlled release and delivery of growth factors by hydrogel-based scaffolds and the enhancement of half-life and selectivity of growth factors by a novel approach facilitated by glycosaminoglycans.
Collapse
|
27
|
Napoli E, Borlongan CV. Recent Advances in Stem Cell-Based Therapeutics for Stroke. Transl Stroke Res 2016; 7:452-457. [PMID: 27515852 PMCID: PMC5065756 DOI: 10.1007/s12975-016-0490-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 08/03/2016] [Indexed: 01/27/2023]
Abstract
Regenerative medicine for central nervous system disorders, including stroke, has challenged the non-regenerative capacity of the brain. Among the many treatment strategies tailored towards repairing the injured brain, stem cell-based therapeutics have been demonstrated as safe and effective in animal models of stroke, and are being tested in limited clinical trials. We address here key lab-to-clinic translational research that relate to efficacy, safety, and mechanism of action underlying stem cell therapy. Recognizing the multi-pronged cell death processes associated with stroke that will likely require combination therapies, we next discuss potent drugs and novel technologies directed at improving the functional outcomes of stem cell-based therapeutics. We also examine discrepant transplant regimens between preclinical studies and clinical trials, as well as missing appropriate control arm (i.e., stroke subjects undergoing rehabilitation) on which to directly compare the therapeutic benefits of cell therapy. Finally, the bioethics of cell therapy is presented in order to assess its prevailing social status. With preliminary results now being reported from on-going clinical trials of stem cell therapy for stroke, a careful assessment of the true functional benefits of this novel treatment will further direct the future of regenerative medicine for neurological disorders.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, University of California Davis, Davis, CA, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
28
|
Min JW, Hu JJ, He M, Sanchez RM, Huang WX, Liu YQ, Bsoul NB, Han S, Yin J, Liu WH, He XH, Peng BW. Vitexin reduces hypoxia-ischemia neonatal brain injury by the inhibition of HIF-1alpha in a rat pup model. Neuropharmacology 2015; 99:38-50. [PMID: 26187393 DOI: 10.1016/j.neuropharm.2015.07.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 06/16/2015] [Accepted: 07/07/2015] [Indexed: 01/25/2023]
Abstract
Previous studies have demonstrated that the early suppression of HIF-1α after hypoxia-ischemia (HI) injury provides neuroprotection. Vitexin (5, 7, 4-trihydroxyflavone-8-glucoside), an HIF-1α inhibitor, is a c-glycosylated flavone that has been identified in medicinal plants. Therefore, we hypothesized that treatment with vitexin would protect against HI brain injury. Newborn rat pups were subjected to unilateral carotid artery ligation followed by 2.5 h of hypoxia (8% O2 at 37 °C). Vitexin (30, 45 or 60 mg/kg) was administered intraperitoneally at 5 min or 3 h after HI. Vitexin, administered 5 min after HI, was neuroprotective as seen by decreased infarct volume evaluated at 48 h post-HI. This neuroprotection was removed when vitexin was administered 3 h after HI. Neuronal cell death, blood-brain barrier (BBB) integrity, brain edema, HIF-1α and VEGF protein levels were evaluated using a combination of Nissl staining, IgG staining, brain water content, immunohistochemistry and Western blot at 24 and 48 h after HI. The long-term effects of vitexin were evaluated by brain atrophy measurement, Nissl staining and neurobehavioral tests. Vitexin (45 mg/kg) ameliorated brain edema, BBB disruption and neuronal cell death; Upregulation of HIF-1α by dimethyloxalylglycine (DMOG) increased the BBB permeability and brain edema compared to HI alone. Vitexin attenuated the increase in HIF-1α and VEGF. Vitexin also had long-term effects of protecting against the loss of ipsilateral brain and improveing neurobehavioral outcomes. In conclusion, our data indicate early HIF-1α inhibition with vitexin provides both acute and long-term neuroprotection in the developing brain after neonatal HI injury.
Collapse
Affiliation(s)
- Jia-Wei Min
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jiang-Jian Hu
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Miao He
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Russell M Sanchez
- Department of Surgery, College of Medicine, Texas A&M Health Science Center, Neuroscience Institute, Scott & White Hospital, & Central Texas Veterans Health Care System, Temple, TX, USA
| | - Wen-Xian Huang
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yu-Qiang Liu
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Najeeb Bassam Bsoul
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Song Han
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jun Yin
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Wan-Hong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xiao-Hua He
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China.
| | - Bi-Wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
29
|
Reis C, Wang Y, Akyol O, Ho WM, Ii RA, Stier G, Martin R, Zhang JH. What's New in Traumatic Brain Injury: Update on Tracking, Monitoring and Treatment. Int J Mol Sci 2015; 16:11903-65. [PMID: 26016501 PMCID: PMC4490422 DOI: 10.3390/ijms160611903] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/04/2015] [Accepted: 05/06/2015] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI), defined as an alteration in brain functions caused by an external force, is responsible for high morbidity and mortality around the world. It is important to identify and treat TBI victims as early as possible. Tracking and monitoring TBI with neuroimaging technologies, including functional magnetic resonance imaging (fMRI), diffusion tensor imaging (DTI), positron emission tomography (PET), and high definition fiber tracking (HDFT) show increasing sensitivity and specificity. Classical electrophysiological monitoring, together with newly established brain-on-chip, cerebral microdialysis techniques, both benefit TBI. First generation molecular biomarkers, based on genomic and proteomic changes following TBI, have proven effective and economical. It is conceivable that TBI-specific biomarkers will be developed with the combination of systems biology and bioinformation strategies. Advances in treatment of TBI include stem cell-based and nanotechnology-based therapy, physical and pharmaceutical interventions and also new use in TBI for approved drugs which all present favorable promise in preventing and reversing TBI.
Collapse
Affiliation(s)
- Cesar Reis
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - Yuechun Wang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
- Department of Physiology, School of Medicine, University of Jinan, Guangzhou 250012, China.
| | - Onat Akyol
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
| | - Wing Mann Ho
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, University Hospital Innsbruck, Tyrol 6020, Austria.
| | - Richard Applegate Ii
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - Gary Stier
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - Robert Martin
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - John H Zhang
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| |
Collapse
|
30
|
Lee JH, Zhang J, Wei L, Yu SP. Neurodevelopmental implications of the general anesthesia in neonate and infants. Exp Neurol 2015; 272:50-60. [PMID: 25862287 DOI: 10.1016/j.expneurol.2015.03.028] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/26/2015] [Accepted: 03/31/2015] [Indexed: 12/17/2022]
Abstract
Each year, about six million children, including 1.5 million infants, in the United States undergo surgery with general anesthesia, often requiring repeated exposures. However, a crucial question remains of whether neonatal anesthetics are safe for the developing central nervous system (CNS). General anesthesia encompasses the administration of agents that induce analgesic, sedative, and muscle relaxant effects. Although the mechanisms of action of general anesthetics are still not completely understood, recent data have suggested that anesthetics primarily modulate two major neurotransmitter receptor groups, either by inhibiting N-methyl-D-aspartate (NMDA) receptors, or conversely by activating γ-aminobutyric acid (GABA) receptors. Both of these mechanisms result in the same effect of inhibiting excitatory activity of neurons. In developing brains, which are more sensitive to disruptions in activity-dependent plasticity, this transient inhibition may have longterm neurodevelopmental consequences. Accumulating reports from preclinical studies show that anesthetics in neonates cause cellular toxicity including apoptosis and neurodegeneration in the developing brain. Importantly, animal and clinical studies indicate that exposure to general anesthetics may affect CNS development, resulting in long-lasting cognitive and behavioral deficiencies, such as learning and memory deficits, as well as abnormalities in social memory and social activity. While the casual relationship between cellular toxicity and neurological impairments is still not clear, recent reports in animal experiments showed that anesthetics in neonates can affect neurogenesis, which could be a possible mechanism underlying the chronic effect of anesthetics. Understanding the cellular and molecular mechanisms of anesthetic effects will help to define the scope of the problem in humans and may lead to preventive and therapeutic strategies. Therefore, in this review, we summarize the current evidence on neonatal anesthetic effects in the developmental CNS and discuss how factors influencing these processes can be translated into new therapeutic strategies.
Collapse
Affiliation(s)
- Jin Hwan Lee
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - James Zhang
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA; Center for Visual and Neurocognitive Rehabilitation, VA Medical Center, Atlanta, GA 30033, USA.
| |
Collapse
|
31
|
Effect of intermittent hypoxia on neuro-functional recovery post brain ischemia in mice. J Mol Neurosci 2014; 55:923-30. [PMID: 25344154 DOI: 10.1007/s12031-014-0447-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/13/2014] [Indexed: 12/28/2022]
Abstract
Intermittent hypoxia was a simulation of a high-altitude environment. Neuro-inflammation post brain ischemia was considered as a vital impact which contributed to cognitive-functional deficit. The isoform of nitric oxide synthase (iNOS) was an inflammation factor secreted by microglias in neuro-inflammation. In this study, we established a high-altitude environment as the hypoxic condition. Twenty mice were selected and randomized into a hypoxia group (n = 10) or a normoxia group (n = 10) post three vessel occlusion-induced brain ischemia. An enhancement of cognitive-functional recovery was presented in the hypoxia group by survival neuron counting and revealed by the Morris water maze test. Meanwhile, a high level of hypoxia-inducable factor 1 (HIF-1) expression associated with a lower expression of iNOS was observed in the border between infarcts and normal tissue of the hippocampus in the hypoxia group. However, these phenomenons were blocked by HIF-1 inhibition. This suggested that the acceleration of cognitive-functional recovery induced by intermittent hypoxia may depend on HIF-1 activating. An imitation of the hypoxic condition with or without HIF-1 inhibition was operated on the BV-2 cell. A high level of HIF-1 expression associated with a lower-level expression of iNOS was performed in the hypoxic condition. These data suggested that intermittent hypoxia can accelerate cognitive function recovery through attenuating neuro-inflammation.
Collapse
|