1
|
Sharma V, Singh TG. Hypoxia-inducible Factor-1α Pathway in Cerebral Ischemia: From Molecular Mechanisms to Therapeutic Targets. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:208-218. [PMID: 39428931 DOI: 10.2174/0118715273324551241008111827] [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: 05/31/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Ischemic injury to the brain can result in a variety of life-threatening conditions, mortality, or varying degrees of disability. Hypoxia-inducible factor 1α (HIF 1α), an oxygen- sensitive transcription factor that controls the adaptive metabolic response to hypoxia, is a critical constituent of cerebral ischemia. It participates in numerous processes, such as metabolism, proliferation, and angiogenesis, and plays a major role in cerebral ischemia. METHODS Through the use of a number of different search engines like Scopus, PubMed, Bentham, and Elsevier databases, a literature review was carried out for investigating the pharmacological modulation of HIF-1α pathways for the treatment of cerebral ischemia. RESULTS Various signalling pathways, such as Mitogen-activated protein kinase (MAPK), Janus kinase/ signal transducers and activators (JAK/STAT), Phosphoinositide-3-kinase (PI3-K), and cAMPresponse element binding protein (CREB) play a vital role in modulation of HIF-1α pathway, which helps in preventing the pathogenesis of cerebral ischemia. CONCLUSION The pharmacological modulation of the HIF-1α pathway via various molecular signalling pathways, such as PI3-K, MAPK, CREB, and JAK/STAT activators, offer a promising prospect for future interventions and treatment for cerebral ischemia.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | |
Collapse
|
2
|
Xie J, Zhang Z. Recent Advances and Therapeutic Implications of 2-Oxoglutarate-Dependent Dioxygenases in Ischemic Stroke. Mol Neurobiol 2024; 61:3949-3975. [PMID: 38041714 DOI: 10.1007/s12035-023-03790-1] [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: 08/04/2023] [Accepted: 11/08/2023] [Indexed: 12/03/2023]
Abstract
Ischemic stroke is a common disease with a high disability rate and mortality, which brings heavy pressure on families and medical insurance. Nowadays, the golden treatments for ischemic stroke in the acute phase mainly include endovascular therapy and intravenous thrombolysis. Some drugs are used to alleviate brain injury in patients with ischemic stroke, such as edaravone and 3-n-butylphthalide. However, no effective neuroprotective drug for ischemic stroke has been acknowledged. 2-Oxoglutarate-dependent dioxygenases (2OGDDs) are conserved and common dioxygenases whose activities depend on O2, Fe2+, and 2OG. Most 2OGDDs are expressed in the brain and are essential for the development and functions of the brain. Therefore, 2OGDDs likely play essential roles in ischemic brain injury. In this review, we briefly elucidate the functions of most 2OGDDs, particularly the effects of regulations of 2OGDDs on various cells in different phases after ischemic stroke. It would also provide promising potential therapeutic targets and directions of drug development for protecting the brain against ischemic injury and improving outcomes of ischemic stroke.
Collapse
Affiliation(s)
- Jian Xie
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
3
|
Chen LH, Hu JN. Development of nano-delivery systems for loaded bioactive compounds: using molecular dynamics simulations. Crit Rev Food Sci Nutr 2024; 65:1811-1832. [PMID: 38206576 DOI: 10.1080/10408398.2023.2301427] [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] [Indexed: 01/12/2024]
Abstract
Over the past decade, a remarkable surge in the development of functional nano-delivery systems loaded with bioactive compounds for healthcare has been witnessed. Notably, the demanding requirements of high solubility, prolonged circulation, high tissue penetration capability, and strong targeting ability of nanocarriers have posed interdisciplinary research challenges to the community. While extensive experimental studies have been conducted to understand the construction of nano-delivery systems and their metabolic behavior in vivo, less is known about these molecular mechanisms and kinetic pathways during their metabolic process in vivo, and lacking effective means for high-throughput screening. Molecular dynamics (MD) simulation techniques provide a reliable tool for investigating the design of nano-delivery carriers encapsulating these functional ingredients, elucidating the synthesis, translocation, and delivery of nanocarriers. This review introduces the basic MD principles, discusses how to apply MD simulation to design nanocarriers, evaluates the ability of nanocarriers to adhere to or cross gastrointestinal mucosa, and regulates plasma proteins in vivo. Moreover, we presented the critical role of MD simulation in developing delivery systems for precise nutrition and prospects for the future. This review aims to provide insights into the implications of MD simulation techniques for designing and optimizing nano-delivery systems in the healthcare food industry.
Collapse
Affiliation(s)
- Li-Hang Chen
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Jiang-Ning Hu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
4
|
A R N, G K R. A deep learning and docking simulation-based virtual screening strategy enables the rapid identification of HIF-1α pathway activators from a marine natural product database. J Biomol Struct Dyn 2024; 42:629-651. [PMID: 37038705 DOI: 10.1080/07391102.2023.2194997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/17/2023] [Indexed: 04/12/2023]
Abstract
Artificial Intelligence is hailed as a cutting-edge technology for accelerating drug discovery efforts, and our goal was to validate its potential in predicting pharmacological inhibitors of EGLN1 using a deep learning-based architecture, one of its subsidiaries. Egl nine homolog 1 (EGLN1) inhibition prevents poly ubiquitination-mediated proteosomal destruction HIF-1α. The pharmacological interventions aimed at stabilizing HIF-1α have the potential to be a promising treatment option for a range of human diseases, including ischemic stroke. To unveil a novel EGLN1 inhibitor from marine natural products, a custom-based virtual screening was carried out using a Deep Convolutional Neural Network (DCNN) architecture, docking, and molecular dynamics simulation. The custom DCNN model was optimized and further employed to screen marine natural products from the CMNPD database. The docking was performed as a secondary strategy for screened hits. Molecular dynamics (MD) and molecular mechanics/generalized Born surface area (MM-GBSA) were used to analyze inhibitor binding and identify key interactions. The findings support the claim that deep learning-based virtual screening is a rapid, reliable and accurate method of identifying highly contributing drug candidates (EGLN1 inhibitors). This study demonstrates that deep learning architecture can significantly accelerate drug discovery and development, and provides a solid foundation for using (Z)-2-ethylhex-2-enedioic acid [(Z)-2-ethylhex-2-enedioic acid] as a potential EGLN1 inhibitor for treating various health complications.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Neelakandan A R
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| | - Rajanikant G K
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| |
Collapse
|
5
|
Kakhki S, Goodarzi M, Abbaszade-Cheragheali A, Rajabi M, Masoumipour AH, Khatibi SR, Beheshti F. Folic acid supplementation improved cognitive deficits associated with lithium administration during pregnancy in rat offspring. Int J Dev Neurosci 2023; 83:615-630. [PMID: 37582655 DOI: 10.1002/jdn.10289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/02/2023] [Accepted: 07/14/2023] [Indexed: 08/17/2023] Open
Abstract
INTRODUCTION The present study aimed to analyse both neurobehavioural and biochemical results of neonates born of mothers exposed to different doses of lithium along with the groups that received lithium at the highest dose with folic acid as a preventive treatment. MATERIALS AND METHODS Male and female rats were mated in separate cages, and pregnant rats were divided into eight first group as (1) vehicle; (2) propylthiouracil (PTU)-induced hypothyroidism; (3-4) received two different doses of lithium carbonate (15 and 30 mg/kg); (5-7) the highest doses of lithium (30 mg/kg) plus three different doses of folic acid (5, 10 and 15 mg/kg); and (8) received just folic acid (15 mg/kg). All treatments were dissolved in drinking water and continued until delivery, followed by returning to a regular diet without treatment. RESULTS Lithium (30 mg/kg) disrupts both behavioural and biochemical markers, including TSH, T3 and T4 as measuring indicators to assess thyroid function, IL-10 and TNF-α as anti-inflammatory and inflammatory agents, respectively, malondialdehyde as an oxidative stress marker, alongside SOD, and catalase activity as antioxidant indicators. Besides, folic acid, almost at the highest dose (15 mg/kg), attenuated memory impairement and anxiety-like behaviour caused by lithium. Moreover, the groups treated with folic acid alone in comparison with vehicles demonstrated higher levels of antioxidant and anti-inflammatory indicators. CONCLUSION According to the results, prenatal exposure to a high dose of lithium (30 mg/kg) leads to foetal neurodevelopmental disorder and growth restriction through various mechanisms more likely attributed to hypothyroidism, which means it should be either prohibited or prescribed cautiously during pregnancy.
Collapse
Affiliation(s)
- Samaneh Kakhki
- Department of Clinical Biochemistry, School of Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mehrnoush Goodarzi
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Ali Abbaszade-Cheragheali
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mojgan Rajabi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Hossein Masoumipour
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Seyed Reza Khatibi
- Department of Public Health, School of Health, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Farimah Beheshti
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
- Departments of Physiology, School of Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| |
Collapse
|
6
|
Vatte S, Ugale R. HIF-1, an important regulator in potential new therapeutic approaches to ischemic stroke. Neurochem Int 2023; 170:105605. [PMID: 37657765 DOI: 10.1016/j.neuint.2023.105605] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023]
Abstract
Ischemic stroke is a leading cause of disability and mortality worldwide due to the narrow therapeutic window of the only approved therapies like intravenous thrombolysis and thrombectomy. Hypoxia inducible factor-1α (HIF-1α) is a sensitive regulator of oxygen homeostasis, and its expression is rapidly induced after hypoxia/ischemia. It plays an extensive role in the pathophysiology of stroke by regulating multiple pathways including glucose metabolism, angiogenesis, neuronal survival, neuroinflammation and blood brain barrier regulation. Here, we give a brief overview of the HIF-1α-targeting strategies currently under investigation and summarise recent research on how HIF-1α is regulated in various brain cells, including neurons and microglia, at various stages in ischemic stroke. The roles of HIF-1 in stroke varies with ischemic time and degree of ischemia, are still up for debate. More focus has been placed on prospective HIF-1α targeting drugs, such as HIF-1α activator, HIF-1α stabilizers, and natural compounds. In this review, we have highlighted the regulation of HIF-1α in the novel therapeutic approaches for treatment of stroke.
Collapse
Affiliation(s)
- Sneha Vatte
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, 440033, India.
| | - Rajesh Ugale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, 440033, India.
| |
Collapse
|
7
|
Dong H, Zhang C, Shi D, Xiao X, Chen X, Zeng Y, Li X, Xie R. Ferroptosis related genes participate in the pathogenesis of spinal cord injury via HIF-1 signaling pathway. Brain Res Bull 2023; 192:192-202. [PMID: 36414158 DOI: 10.1016/j.brainresbull.2022.11.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 10/29/2022] [Accepted: 11/17/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Spinal cord injury (SCI) is a crushing disease without a effective and specific therapeutic strategy. Therefore, it is crucial to uncover underlying mechanism in order to identify potential treatments for SCI. Current studies show ferroptosis might pay important role in SCI. METHODS In this study, we aimed to identify the key ferroptosis-related genes providing therapeutic targets for SCI. GSE45006, GSE19890 and GSE156999 from Gene Expression Omnibus (GEO) database were analyzed. RESULTS A total of 61 ferroptosis-related DEGs were identified, followed by bioinformatics enrichment analyses and PPI network construction. Ten key ferroptosis-related genes were identified by Cytoscape (Cytohubba), most of which were enriched in the HIF-1 signaling pathway. Then we constructed a clip SCI rat model and qPCR was performed to assess the expressions of five genes enriched in HIF-1 signaling pathway (Stat3, Tlr4, Hmox1, Hif1a and Cybb). Finally, a ceRNA network, Stat3, Tlr4, Hmox1/miR127, miR383, miR485/rno-Mut_0003, rno-Pwwp2a_0002 was constructed and expression of mentioned molecules were validated by chip data. CONCLUSIONS Five hub genes from HIF-1 signaling pathway were identified and might play a central role in SCI, which indicated that ferroptosis was correlated with HIF-1 signaling pathway. These results can provide a new insight into molecular mechanisms and identify potential therapeutic targets for SCI.
Collapse
Affiliation(s)
- Haoru Dong
- Department of Neurosurgery; National Center for Neurological Disorders; Neurosurgical Institute of Fudan University; Shanghai Clinical Medical Center of Neurosurgery; Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Chi Zhang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Donglei Shi
- Department of Nursing, Huashan Hospital, Fudan University, Shanghai 200032, China.
| | - Xiao Xiao
- Department of Neurosurgery; National Center for Neurological Disorders; Neurosurgical Institute of Fudan University; Shanghai Clinical Medical Center of Neurosurgery; Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Xingyu Chen
- Department of Neurosurgery; National Center for Neurological Disorders; Neurosurgical Institute of Fudan University; Shanghai Clinical Medical Center of Neurosurgery; Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Yuanxiao Zeng
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xiaomu Li
- Department of Endocrinology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Rong Xie
- Department of Neurosurgery; National Center for Neurological Disorders; Neurosurgical Institute of Fudan University; Shanghai Clinical Medical Center of Neurosurgery; Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Neurosurgery, National Regional Medical Center; Huashan Hospital Fujian Campus, Fudan University; The First Affiliated Hospital of Fujian Medical University, Fuzhou 350209, Fujian Province, China.
| |
Collapse
|
8
|
Das J, Mahammad FS, Krishnamurthy RG. An integrated chemo-informatics and in vitro experimental approach repurposes acarbose as a post-ischemic neuro-protectant. 3 Biotech 2022; 12:71. [PMID: 35223357 PMCID: PMC8847516 DOI: 10.1007/s13205-022-03130-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 01/23/2022] [Indexed: 11/26/2022] Open
Abstract
The increasing prevalence of ischemic stroke combined with limited therapeutic options highlights the compelling need for continued research into the development of future neuro-therapeutics. Death-Associated Protein Kinase 1 (DAPK1) and p53 protein-protein interaction serve as a signaling point for the convergence of apoptosis and necrosis in cerebral ischemia. In this study, we used an integrated chemo-informatics and in vitro experimental drug repurposing strategy to screen potential small-molecule inhibitors of DAPK1-p53 interaction from the United States of America Food and Drug Administration (FDA) approved drug database exhibiting post-ischemic neuroprotective and neuro-regenerative efficacy and mechanisms. The computational docking and molecular dynamics simulation of FDA-approved drugs followed by an in vitro experimental validation identified acarbose, an anti-diabetic medication and caloric restriction mimetic as a potential inhibitor of DAPK1-p53 interaction. The evaluation of post-ischemic neuroprotective and regenerative efficacy and mechanisms of action for acarbose was carried out using a set of experimental methods, including cell viability, proliferation and differentiation assays, fluorescence staining, and gene expression analysis. Post-ischemic administration of acarbose conferred significant neuroprotection against ischemia-reperfusion injury in vitro. The reduced fluorescence emission in cells stained with pS20 supported the potential of acarbose in inhibiting the DAPK1-p53 interaction. Acarbose prevented mitochondrial and lysosomal dysfunction, and favorably modulated gene expression related to cell survival, inflammation, and regeneration. BrdU staining and neurite outgrowth assay showed a significant increase in cell proliferation and differentiation in acarbose-treated group. This is the first study known to provide mechanistic insight into the post-ischemic neuroprotective and neuro-regenerative potential of acarbose. Our results provide a strong basis for preclinical studies to evaluate the safety and neuroprotective efficacy of acarbose against ischemic stroke. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13205-022-03130-5.
Collapse
Affiliation(s)
- Jyotirekha Das
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala 673601 India
| | - Fayaz Shaik Mahammad
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka 576104 India
| | | |
Collapse
|
9
|
Pan Z, Ma G, Kong L, Du G. Hypoxia-inducible factor-1: Regulatory mechanisms and drug development in stroke. Pharmacol Res 2021; 170:105742. [PMID: 34182129 DOI: 10.1016/j.phrs.2021.105742] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/13/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022]
Abstract
Stroke is an acute cerebrovascular disease caused by sudden rupture of blood vessels in the brain or blockage of blood vessels, which has now become one of the main causes of adult death. During stroke, hypoxia-inducible factor-1 (HIF-1), as an important regulator under hypoxia conditions, is involved in the pathological process of stroke by regulating multi-pathways, such as glucose metabolism, angiogenesis, erythropoiesis, cell survival. However, the roles of HIF-1 in stroke are still controversial, which are related with ischemic time and degree of ischemia. The regulatory mechanisms of HIF-1 in stroke include inflammation, autophagy, oxidative stress, apoptosis and energy metabolism. The potential drugs targeting HIF-1 have attracted more attention, such as HIF-1 inhibitors, HIF-1 stabilizers and natural products. Based on the role of HIF-1 in stroke, HIF-1 is expected to be a potential target for stroke treatment. Resolving when and what interventions for HIF-1 to take during stroke will provide novel strategies for stroke treatment.
Collapse
Affiliation(s)
- Zirong Pan
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Guodong Ma
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Linglei Kong
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| | - Guanhua Du
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
10
|
Wu Y, Li Z, McDonough MA, Schofield CJ, Zhang X. Inhibition of the Oxygen-Sensing Asparaginyl Hydroxylase Factor Inhibiting Hypoxia-Inducible Factor: A Potential Hypoxia Response Modulating Strategy. J Med Chem 2021; 64:7189-7209. [PMID: 34029087 DOI: 10.1021/acs.jmedchem.1c00415] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Factor inhibiting hypoxia-inducible factor (FIH) is a JmjC domain 2-oxogluarate and Fe(II)-dependent oxygenase that catalyzes hydroxylation of specific asparagines in the C-terminal transcriptional activation domain of hypoxia-inducible factor alpha (HIF-α) isoforms. This modification suppresses the transcriptional activity of HIF by reducing its interaction with the transcriptional coactivators p300/CBP. By contrast with inhibition of the HIF prolyl hydroxylases (PHDs), inhibitors of FIH, which accepts multiple non-HIF substrates, are less studied; they are of interest due to their potential ability to alter metabolism (either in a HIF-dependent and/or -independent manner) and, provided HIF is upregulated, to modulate the course of the HIF-mediated hypoxic response. Here we review studies on the mechanism and inhibition of FIH. We discuss proposed biological roles of FIH including its regulation of HIF activity and potential roles of FIH-catalyzed oxidation of non-HIF substrates. We highlight potential therapeutic applications of FIH inhibitors.
Collapse
Affiliation(s)
- Yue Wu
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Zhihong Li
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Michael A McDonough
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Xiaojin Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
11
|
Burgess K, Bennett C, Mosnier H, Kwatra N, Bethel F, Jadavji NM. The Antioxidant Role of One-Carbon Metabolism on Stroke. Antioxidants (Basel) 2020; 9:E1141. [PMID: 33212887 PMCID: PMC7698340 DOI: 10.3390/antiox9111141] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
One-carbon (1C) metabolism is a metabolic network that is centered on folate, a B vitamin; it integrates nutritional signals with biosynthesis, redox homeostasis, and epigenetics. This metabolic pathway also reduces levels of homocysteine, a non-protein amino acid. High levels of homocysteine are linked to increased risk of hypoxic events, such as stroke. Several preclinical studies have suggested that 1C metabolism can impact stroke outcome, but the clinical data are unclear. The objective of this paper was to review preclinical and clinical research to determine whether 1C metabolism has an antioxidant role on stroke. To accomplish the objective, we searched for publications using the following medical subject headings (MeSH) keywords: antioxidants, hypoxia, stroke, homocysteine, one-carbon metabolism, folate, methionine, and dietary supplementation of one-carbon metabolism. Both pre-clinical and clinical studies were retrieved and reviewed. Our review of the literature suggests that deficiencies in 1C play an important role in the onset and outcome of stroke. Dietary supplementation of 1C provides beneficial effects on stroke outcome. For stroke-affected patients or individuals at high risk for stroke, the data suggest that nutritional modifications in addition to other therapies could be incorporated into a treatment plan.
Collapse
Affiliation(s)
- Kassidy Burgess
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA;
- Biomedical Sciences Program, Midwestern University, Glendale, AZ 85308, USA; (C.B.); (N.K.); (F.B.)
| | - Calli Bennett
- Biomedical Sciences Program, Midwestern University, Glendale, AZ 85308, USA; (C.B.); (N.K.); (F.B.)
- College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Hannah Mosnier
- School of Medicine, National University of Ireland Galway, H91 TK33, Ireland;
- College of Dental Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Neha Kwatra
- Biomedical Sciences Program, Midwestern University, Glendale, AZ 85308, USA; (C.B.); (N.K.); (F.B.)
- College of Dental Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Forrest Bethel
- Biomedical Sciences Program, Midwestern University, Glendale, AZ 85308, USA; (C.B.); (N.K.); (F.B.)
- College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Nafisa M. Jadavji
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA;
- Biomedical Sciences Program, Midwestern University, Glendale, AZ 85308, USA; (C.B.); (N.K.); (F.B.)
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
12
|
Folic Acid Deficiency Enhances the Tyr705 and Ser727 Phosphorylation of Mitochondrial STAT3 in In Vivo and In Vitro Models of Ischemic Stroke. Transl Stroke Res 2020; 12:829-843. [PMID: 33037575 DOI: 10.1007/s12975-020-00860-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 01/01/2023]
Abstract
Ischemic stroke remains one of the most common causes of death and disability worldwide. The stroke patients with an inadequate intake of folic acid tend to have increased brain injury and poorer prognosis. However, the precise mechanisms underlying the harmful effects of folic acid deficiency (FD) in ischemic stroke is still elusive. Here, we aimed to test the hypothesis that mitochondrial localized STAT3 (mitoSTAT3) expression may be involved in the process of neuronal damage induced by FD in in vivo and in vitro models of ischemic stroke. Our results exhibited that FD increased infarct size and aggravated the damage of mitochondrial ultrastructure in ischemic brains. Meanwhile, FD upregulated the phosphorylation levels of mitoSTAT3 at Tyr705 (Y705) and Ser727 (S727) sites in the rat middle cerebral artery occlusion/reperfusion (MCAO/R) model and oxygen-glucose deprivation followed by reperfusion (OGD/R) N2a cells. Furthermore, the inhibition of JAK2 by AG490 led to a significant decrease in FD-induced phosphorylation of Y705, while S727 phosphorylation was unaffected. Conversely, U0126 and LY294002, which respectively inhibited phosphorylation of ERK1/2 and Akt, partially prevented S727 phosphorylation, but had limited effects on the level of pY705, suggesting that phosphorylation of Y705 and S727 is regulated via independent mechanisms in FD-treated brains.
Collapse
|
13
|
Postischemic supplementation of folic acid improves neuronal survival and regeneration in vitro. Nutr Res 2020; 75:1-14. [DOI: 10.1016/j.nutres.2019.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/05/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023]
|
14
|
Li Y, Han W, Wu Y, Zhou K, Zheng Z, Wang H, Xie L, Li R, Xu K, Liu Y, Wang X, Xiao J. Stabilization of Hypoxia Inducible Factor-1α by Dimethyloxalylglycine Promotes Recovery from Acute Spinal Cord Injury by Inhibiting Neural Apoptosis and Enhancing Axon Regeneration. J Neurotrauma 2019; 36:3394-3409. [PMID: 31232175 DOI: 10.1089/neu.2018.6364] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Yao Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wen Han
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanqing Wu
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang, China
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhilong Zheng
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haoli Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ling Xie
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rui Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ke Xu
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang, China
| | - Yanlong Liu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Xiao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
15
|
Cai D, Qi J, Yang Y, Zhang W, Zhou F, Jia X, Guo W, Huang X, Gao F, Chen H, Li T, Li G, Wang P, Zhang Y, Lei H. Design, Synthesis and Biological Evaluation of Diosgenin-Amino Acid Derivatives with Dual Functions of Neuroprotection and Angiogenesis. Molecules 2019; 24:molecules24224025. [PMID: 31703284 PMCID: PMC6891328 DOI: 10.3390/molecules24224025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 12/20/2022] Open
Abstract
Diosgenin, a natural product with steroidal structure, has a wide range of clinical applications in China. It also shows great potential in the treatment of blood clots and nerve damage. To enhance the bioavailability as well as efficacy of diosgenin, eighteen diosgenin-amino acid derivatives were designed and synthesized. The neuroprotective effects of these compounds were evaluated by SH-SY5Y cell line and the biosafety was evaluated by H9c2 cell line. The results displayed that part of the derivatives' activities (EC50 < 20 μM) were higher than positive control edaravone (EC50 = 21.60 ± 3.04 μM), among which, DG-15 (EC50 = 6.86 ± 0.69 μM) exhibited the best neuroprotection. Meanwhile, biosafety evaluation showed that DG-15 had no cytotoxicity on H9c2 cell lines. Interestingly, combined neuroprotective and cytotoxic results, part of the derivatives without their protecting group were superior to compounds with protecting group. Subsequently, Giemsa staining and DAPI (4',6-diamidino-2-phenylindole) staining indicated that DG-15 had a protective effect on damaged SH-SY5Y cells by reducing apoptosis. Moreover, DG-15 showed a higher role in promoting angiogenesis at high concentrations (4 mg/mL) on the chorioallantoic membrane model. This finding displayed that DG-15 had dual functions of neuroprotection and angiogenesis, which provided further insight into designing agent for the application in treatment of ischemic stroke.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Yuzhong Zhang
- Correspondence: (Y.Z.); (H.L.); Tel.: +86-10-8473-8645 (H.L.)
| | - Haimin Lei
- Correspondence: (Y.Z.); (H.L.); Tel.: +86-10-8473-8645 (H.L.)
| |
Collapse
|
16
|
Mutavdzin S, Gopcevic K, Stankovic S, Jakovljevic Uzelac J, Labudovic Borovic M, Djuric D. The effect of folic acid administration on cardiac tissue matrix metalloproteinase activity and hepatorenal biomarkers in diabetic rats 1. Can J Physiol Pharmacol 2019; 97:893-901. [PMID: 31295411 DOI: 10.1139/cjpp-2019-0027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Diabetes mellitus (DM) is a metabolic disorder that causes severe complications. Thus, the aims of this study were to investigate the influence of DM and folic acid treatment on liver and renal biomarkers, and heart remodeling through evaluation of cardiac matrix metalloproteinase (MMP) activity. There were 4 groups: control (physiological saline 1 mL/kg, i.p., 28 days), DM (streptozotocin [STZ] 100 mg/kg in physiological saline, i.p., 1 day), folic acid (FA; 5 mg/kg, i.p., 28 days), and DM+FA (STZ 100 mg/kg, i.p., 1 day and folic acid 5 mg/kg, i.p., 28 days). Our results demonstrated increased aminotransferase and alkaline phosphatase activity, urea and creatinine concentration, and decreased albumin and fibrinogen concentration in the DM group. MMP-2 relative activity was elevated in the DM and FA groups; MMP-9 was decreased in the DM and increased in the FA group. The folic acid treatment of diabetic rats did not change aminotransferase activity; it alleviated the increase in alkaline phosphatase and the decrease in albumin and fibrinogen concentration, and reduced MMP-2 activity; however, it increased urea and creatinine concentration. In conclusion, folic acid treatment of diabetic rats has cardio- and hepato-protective effects. However, its dosing should be carefully considered because of possible renal damage.
Collapse
Affiliation(s)
- Slavica Mutavdzin
- Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Kristina Gopcevic
- Institute of Chemistry in Medicine "Prof. Dr. Petar Matavulj", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Sanja Stankovic
- Centre of Medical Biochemistry, Clinical Centre of Serbia, Belgrade, Serbia
| | - Jovana Jakovljevic Uzelac
- Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milica Labudovic Borovic
- Institute of Histology and Embryology "Aleksandar Dj. Kostic", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dragan Djuric
- Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
17
|
Nampoothiri SS, Rajanikant GK. miR-9 Upregulation Integrates Post-ischemic Neuronal Survival and Regeneration In Vitro. Cell Mol Neurobiol 2019; 39:223-240. [PMID: 30539420 PMCID: PMC11469860 DOI: 10.1007/s10571-018-0642-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 12/07/2018] [Indexed: 01/02/2023]
Abstract
The irrefutable change in the expression of brain-enriched microRNAs (miRNAs) following ischemic stroke has promoted the development of radical miRNA-based therapeutics encompassing neuroprotection and neuronal restoration. Our previous report on the systems-level prediction of miR-9 in post-stroke-induced neurogenesis served as a premise to experimentally uncover the functional role of miR-9 in post-ischemic neuronal survival and regeneration. The oxygen-glucose deprivation (OGD) in SH-SY5Y cells significantly reduced miR-9 expression, while miR-9 mimic transfection enhanced post-ischemic neuronal cell viability. The next major objective involved the execution of a drug repositioning strategy to augment miR-9 expression via structure-based screening of Food and Drug Administration (FDA)-approved drugs that bind to Histone Deacetylase 4 (HDAC4), a known miR-9 target. Glucosamine emerged as the top hit and its binding potential to HDAC4 was verified by Molecular Dynamics (MD) Simulation, Drug Affinity Responsive Target Stability (DARTS) assay, and MALDI-TOF MS. It was intriguing that the glucosamine treatment 1-h post-OGD was associated with the increased miR-9 level as well as enhanced neuronal viability. miR-9 mimic or post-OGD glucosamine treatment significantly increased the cellular proliferation (BrdU assay), while the neurite outgrowth assay displayed elongated neurites. The enhanced BCL2 and VEGF parallel with the reduced NFκB1, TNF-α, IL-1β, and iNOS mRNA levels in miR-9 mimic or glucosamine-treated cells further substantiated their post-ischemic neuroprotective and regenerative efficacy. Hence, this study unleashes a potential therapeutic approach that integrates neuronal survival and regeneration via small-molecule-based regulation of miR-9 favoring long-term recovery against ischemic stroke.
Collapse
Affiliation(s)
- Sreekala S Nampoothiri
- School of Biotechnology, National Institute of Technology Calicut, Calicut, 673601, India
| | - G K Rajanikant
- School of Biotechnology, National Institute of Technology Calicut, Calicut, 673601, India.
| |
Collapse
|
18
|
Davis CK, Jain SA, Bae ON, Majid A, Rajanikant GK. Hypoxia Mimetic Agents for Ischemic Stroke. Front Cell Dev Biol 2019; 6:175. [PMID: 30671433 PMCID: PMC6331394 DOI: 10.3389/fcell.2018.00175] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/10/2018] [Indexed: 12/27/2022] Open
Abstract
Every year stroke claims more than 6 million lives worldwide. The majority of them are ischemic stroke. Small molecule-based therapeutics for ischemic stroke has attracted a lot of attention, but none has been shown to be clinically useful so far. Hypoxia-inducible factor-1 (HIF-1) plays a crucial role in the transcriptional adaptation of cells to hypoxia. Small molecule-based hypoxia-mimetic agents either stabilize HIF-1α via HIF-prolyl hydroxylases (PHDs) inhibition or through other mechanisms. In both the cases, these agents have been shown to confer ischemic neuroprotection in vitro and in vivo. The agents which act via PHD inhibition are mainly classified into iron chelators, iron competitors, and 2 oxoglutarate (2OG) analogs. This review discusses HIF structure and key players in the HIF-1 degradation pathway as well as the genes, proteins and chemical molecules that are connected to HIF-1 and how they affect cell survival following ischemic injury. Furthermore, this review gives a summary of studies that used PHD inhibitors and other HIF-1α stabilizers as hypoxia-mimetic agents for the treatment of ischemic injury.
Collapse
Affiliation(s)
- Charles K. Davis
- School of Biotechnology, National Institute of Technology Calicut, Calicut, India
| | - Saurabh A. Jain
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Ok-Nam Bae
- College of Pharmacy, Hanyang University, Ansan, South Korea
| | - Arshad Majid
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - G. K. Rajanikant
- School of Biotechnology, National Institute of Technology Calicut, Calicut, India
| |
Collapse
|