1
|
Hansda S, Das H. Insights into Cancer-Associated Thrombosis Leading Towards Ischemic Stroke. BIOLOGY 2025; 14:50. [PMID: 39857281 PMCID: PMC11762743 DOI: 10.3390/biology14010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025]
Abstract
Stroke leads to significant disability in most patients, whereas cancer elevates the occurrence of stroke. The incidence of cancer-associated stroke (CAS) is projected to rise as a result of improvements in cancer therapies. Various forms of cancer have been demonstrated to be linked to ischemic stroke. Cancer might influence stroke pathophysiology either directly or through coagulation that creates a hypercoagulative state, in addition to infections. Treatment methods for cancer, including chemotherapy, radiotherapy, and surgery, have all been demonstrated to increase the risk of stroke as well. This review discusses the subtypes, epidemiology, pathophysiology, mechanisms of stroke within cancer patients, biomarkers, and signaling pathways of stroke in cancer while providing vital information on the involved transcription factors, treatment, and management of patients with cancer-associated ischemic stroke. Atherosclerosis, extracellular vesicles (EVs), and signaling biomolecules can also affect CAS. Overall, stroke is a significant and not uncommon complication of cancer, and there is an immediate demand for neurologists and oncologists to create strategies for screening and preventing strokes in cancer patients.
Collapse
Affiliation(s)
| | - Hiranmoy Das
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| |
Collapse
|
2
|
Cao X, Pu Y. Curcumin Regulates Microglia Polarization to Alleviate Ischemic Stroke by Targeting microRNA-205-5p/Kruppel-Like Factor 2 (KLF2)/Activating Transcription Factor 2 (ATF2) Axis. Chem Biol Drug Des 2025; 105:e70050. [PMID: 39821443 DOI: 10.1111/cbdd.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/16/2024] [Accepted: 01/04/2025] [Indexed: 01/19/2025]
Abstract
Ischemic stroke (IS) often causes fearful sequela, even death. Curcumin was beneficial to IS, but its underlying molecular mechanism is unclear. Mice were subjected to middle cerebral artery occlusion (MCAO) surgery, and BV-2 cells were treated with oxygen-glucose deprivation/reoxygenation (OGD/R) induction to establish IS models in vivo and in vitro. Abundance of genes and proteins was determined using quantitative real-time polymerase chain reaction (RT-qPCR), immunofluorescence (IF), and western blot. Interleukin-1β (IL-1β), interleukin-6 (IL-6), and interleukin-10 (IL-10) levels were analyzed using enzyme-linked immunosorbent assay (ELISA). Modified neurological severity score (mNSS), corner test, foot fault test, adhesive removal test, and 2,3,5-triphenyltetrazolium chloride (TTC) staining were applied to evaluate the brain injury of mice. The correlation between miR-205-5p and Kruppel-like factor 2 (KLF2) was affirmed using dual luciferase reporter assay. Our results revealed that curcumin alleviated brain damage in MCAO mice through driving microglia M2 polarization. Of note, curcumin resulted in decreased miR-205-5p expression in MCAO mice. miR-205-5p knockdown resulted in promoted microglia M2 polarization in OGD/R conditions and achieved similar results to curcumin treatment in MCAO mice. Moreover, curcumin played a promoting role in microglia M2 polarization under OGD/R conditions, while miR-205-5p overexpression or KLF2 knockdown abolished these effects. On the mechanism, miR-205-5p was a target of curcumin, and miR-205-5p further interacted with KLF2 to inhibit activating transcription factor 2 (ATF2) expression. miR-205-5p, decreased by curcumin, suppressed microglia M2 polarization to worsen IS injury through the mediating KLF2/ATF2 axis.
Collapse
Affiliation(s)
- Xiangyu Cao
- Hunan University of Medicine, Huaihua, China
| | - Yingzi Pu
- Huaihua Hospital of Traditional Chinese Medicine, Huaihua, China
| |
Collapse
|
3
|
Liu JC, Lei SY, Zhang DH, He QY, Sun YY, Zhu HJ, Qu Y, Zhou SY, Yang Y, Li C, Guo ZN. The pleiotropic effects of statins: a comprehensive exploration of neurovascular unit modulation and blood-brain barrier protection. Mol Med 2024; 30:256. [PMID: 39707228 DOI: 10.1186/s10020-024-01025-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
The blood-brain barrier (BBB) is the most central component of the neurovascular unit (NVU) and is crucial for the maintenance of the internal environment of the central nervous system and the regulation of homeostasis. A multitude of neuroprotective agents have been developed to exert neuroprotective effects and improve the prognosis of patients with ischemic stroke. These agents have been designed to maintain integrity and promote BBB repair. Statins are widely used as pharmacological agents for the treatment and prevention of ischemic stroke, making them a cornerstone in the pharmacological armamentarium for this condition. The primary mechanism of action is the reduction of serum cholesterol through the inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, which results in a decrease in low-density lipoprotein cholesterol (LDL-C) and an increase in cholesterol clearance. Nevertheless, basic and clinical research has indicated that statins may exert additional pleiotropic effects beyond LDL-C reduction. Previous studies on ischemic stroke have demonstrated that statins can enhance neurological function, reduce inflammation, and promote angiogenic and synaptic processes following ischemic stroke. The BBB has been increasingly recognized for its role in the development and progression of ischemic stroke. Statins have also been found to play a potential BBB protective role by affecting members of the NVU. This review aimed to provide a comprehensive theoretical basis for the clinical application of statins by systematically detailing how statins influence the BBB, particularly focusing on the regulation of the function of each member of the NVU.
Collapse
Affiliation(s)
- Jia-Cheng Liu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Shuang-Yin Lei
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Dian-Hui Zhang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Qian-Yan He
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Ying-Ying Sun
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Hong-Jing Zhu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Yang Qu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Sheng-Yu Zhou
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Yi Yang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Xinmin Street 1#, Changchun, 130021, China
| | - Chao Li
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China.
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Xinmin Street 1#, Changchun, 130021, China.
- Neuroscience Research Center, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China.
| |
Collapse
|
4
|
Hansda S, Prateeksha P, Das H. Krüppel-like factor 2 (KLF2), a potential target for neuroregeneration. Neural Regen Res 2024; 19:2327-2328. [PMID: 38526258 PMCID: PMC11090422 DOI: 10.4103/nrr.nrr-d-23-01758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/01/2023] [Accepted: 12/22/2023] [Indexed: 03/26/2024] Open
Affiliation(s)
- Surajit Hansda
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Prateeksha Prateeksha
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Hiranmoy Das
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| |
Collapse
|
5
|
Hoshino T, Takase H, Ishikawa H, Hamanaka G, Kimura S, Fukuda N, Park JH, Nakajima H, Shirakawa H, Shindo A, Kim KW, H Gelman I, Lok J, Arai K. Transcriptomic Profiles of AKAP12 Deficiency in Mouse Corpus Callosum. Bioinform Biol Insights 2024; 18:11779322241276936. [PMID: 39345723 PMCID: PMC11439161 DOI: 10.1177/11779322241276936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/06/2024] [Indexed: 10/01/2024] Open
Abstract
A-kinase anchor protein 12 (AKAP12), a scaffold protein, has been implicated in the central nervous system, including blood-brain barrier (BBB) function. Although its expression level in the corpus callosum is higher than in other brain regions, such as the cerebral cortex, the role of AKAP12 in the corpus callosum remains unclear. In this study, we investigate the impact of AKAP12 deficiency by transcriptome analysis using RNA-sequencing (RNA-seq) on the corpus callosum of AKAP12 knockout (KO) mice. We observed minimal changes, with only 13 genes showing differential expression, including Akap12 itself. Notably, Klf2 and Sgk1, genes potentially involved in BBB function, were downregulated in AKAP12 KO mice and expressed in vascular cells similar to Akap12. These changes in gene expression may affect important biological pathways that may be associated with neurological disorders. Our findings provide an additional data set for future research on the role of AKAP12 in the central nervous system.
Collapse
Affiliation(s)
- Tomonori Hoshino
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Hajime Takase
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- YCU Center for Novel and Exploratory Clinical Trials (Y-NEXT), Yokohama City University Hospital, Yokohama, Japan
| | - Hidehiro Ishikawa
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Gen Hamanaka
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Shintaro Kimura
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Norito Fukuda
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Ji Hyun Park
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Hiroki Nakajima
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Akihiro Shindo
- Department of Neurology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Kyu-Won Kim
- Research Institute of Pharmaceutical Science, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Irwin H Gelman
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Josephine Lok
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Pediatric Critical Care Medicine, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
6
|
Santos AB, Carona A, Ettcheto M, Camins A, Falcão A, Fortuna A, Bicker J. Krüppel-like factors: potential roles in blood-brain barrier dysfunction and epileptogenesis. Acta Pharmacol Sin 2024; 45:1765-1776. [PMID: 38684799 PMCID: PMC11335766 DOI: 10.1038/s41401-024-01285-w] [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: 12/22/2023] [Accepted: 04/07/2024] [Indexed: 05/02/2024] Open
Abstract
Epilepsy is a chronic and debilitating neurological disorder, known for the occurrence of spontaneous and recurrent seizures. Despite the availability of antiseizure drugs, 30% of people with epilepsy experience uncontrolled seizures and drug resistance, evidencing that new therapeutic options are required. The process of epileptogenesis involves the development and expansion of tissue capable of generating spontaneous recurrent seizures, during which numerous events take place, namely blood-brain barrier (BBB) dysfunction, and neuroinflammation. The consequent cerebrovascular dysfunction results in a lower seizure threshold, seizure recurrence, and chronic epilepsy. This suggests that improving cerebrovascular health may interrupt the pathological cycle responsible for disease development and progression. Krüppel-like factors (KLFs) are a family of zinc-finger transcription factors, encountered in brain endothelial cells, glial cells, and neurons. KLFs are known to regulate vascular function and changes in their expression are associated with neuroinflammation and human diseases, including epilepsy. Hence, KLFs have demonstrated various roles in cerebrovascular dysfunction and epileptogenesis. This review critically discusses the purpose of KLFs in epileptogenic mechanisms and BBB dysfunction, as well as the potential of their pharmacological modulation as therapeutic approach for epilepsy treatment.
Collapse
Affiliation(s)
| | - Andreia Carona
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Miren Ettcheto
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Antoni Camins
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Amílcar Falcão
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Ana Fortuna
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Joana Bicker
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal.
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal.
| |
Collapse
|
7
|
Eshraghi R, Sadati S, Bahrami A, Mirjalili SR, Farrokhian A, Mahjoubin-Tehran M, Mirzaei H. Unveiling the role of long non-coding RNA MALAT1: a comprehensive review on myocardial infarction. Front Cardiovasc Med 2024; 11:1429858. [PMID: 39171328 PMCID: PMC11335503 DOI: 10.3389/fcvm.2024.1429858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024] Open
Abstract
Myocardial infarction (MI) stands at top global causes of death in developed countries, owing mostly to atherosclerotic plaque growth and endothelial injury-induced reduction in coronary blood flow. While early reperfusion techniques have improved outcomes, long-term treatment continues to be difficult. The function of lncRNAs extends to regulating gene expression in various conditions, both physiological and pathological, such as cardiovascular diseases. The objective of this research is to extensively evaluate the significance of the lncRNA called Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) in the development and management of MI. According to research, MALAT1 is implicated in processes such as autophagy, apoptosis, cell proliferation, and inflammation in the cardiovascular system. This investigation examines recent research examining the effects of MALAT1 on heart function and its potential as a mean of diagnosis and treatment for post- MI complications and ischemic reperfusion injury.
Collapse
Affiliation(s)
- Reza Eshraghi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sina Sadati
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ashkan Bahrami
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Reza Mirjalili
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alireza Farrokhian
- Department of Cardiology, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Mahjoubin-Tehran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
8
|
Li Y, Liu C, Chen Z, Lin H, Li X. Netrin-1 protects blood-brain barrier (BBB) integrity after cerebral ischemia-reperfusion by activating the Kruppel-like factor 2 (KLF2)/occludin pathway. J Biochem Mol Toxicol 2024; 38:e23623. [PMID: 38229322 DOI: 10.1002/jbt.23623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/04/2023] [Accepted: 12/14/2023] [Indexed: 01/18/2024]
Abstract
Ischemia/reperfusion (I/R)-induced neural damage and neuroinflammation have been associated with pathological progression during stroke. Netrin-1 is an important member of the family of laminin-related secreted proteins, which plays an important role in governing axon elongation. However, it is unknown whether Netrin-1 possesses a beneficial role in stroke. Here, we employed the middle cerebral artery occlusion (MCAO) model to study the function of Netrin-1 in alleviating brain injuries. Our results demonstrate that Netrin-1 rescued poststroke neurological deficits and inhibited production of the inflammatory cytokines such as interleukin 6 (IL-6) and endothelial chemokine (C-X-C motif) ligand 1 (Cxcl1). Importantly, Netrin-1 protected against MCAO-induced dysfunction of the blood-brain barrier (BBB) in mice and a reduction in the expression of the tight junction (TJ) protein occludin. Additionally, we report that Netrin-1 could ameliorate oxygen-glucose deprivation/reoxygenation (OGD/R)-induced injury and prevent aggravation in endothelial monolayer permeability in bEnd.3 human brain microvascular endothelial cells (HBMVECs). Mechanistically, Netrin-1 ameliorated OGD/R-induced decrease in occludin and Kruppel-like factor 2 (KLF2) in HBMVECs. Notably, silencing of KLF2 abolished the beneficial effects of Netrin-1 in protecting endothelial permeability and occludin expression, suggesting that these effects are mediated by KLF2. In conclusion, our findings suggest that Netrin-1 could constitute a novel therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Yuanxiao Li
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Changyun Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Zhiting Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Hanbin Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Xiaofeng Li
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| |
Collapse
|
9
|
Gong F, Shi Q, Mou X, Wang K, Wang Q, Wang H. Atorvastatin mitigates memory deficits and brain monocyte infiltration in chronic hypercholesterolemia. Aging (Albany NY) 2023; 15:13669-13679. [PMID: 38048213 PMCID: PMC10756112 DOI: 10.18632/aging.205217] [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: 04/21/2023] [Accepted: 09/08/2023] [Indexed: 12/06/2023]
Abstract
Mild cognitive impairment (MCI) is a common symptom observed in people over 60 years old and is found to be aggravated by hypercholesterolemia. Severe neuroinflammation induced by BBB dysfunction and monocyte infiltration might be responsible for neuron damage and cognitive impairment. Atorvastatin is a lipid-lowering drug that is widely applied for the treatment of cardiovascular diseases. However, the potential function of Atorvastatin in hypercholesterolemia-induced MCI remains uncertain. Our research will explore the potential therapeutic function of Atorvastatin in memory deficits induced by chronic hypercholesterolemia. ApoE-/- mice were utilized to mimic the state of chronic hypercholesterolemia and were divided into four groups. Animals in the WT and ApoE-/-groups were orally administered with normal saline, while WT mice in the Atorvastatin group and ApoE-/- mice in the ApoE-/-+ Atorvastatin group were orally administered with 10 mg/kg/day Atorvastatin. Markedly increased plasma cholesterol levels reduced RI in the long-term memory test and the spatial short-term memory test, declined mobility in the open field test, and downregulated PSD-95 and BDNF were observed in ApoE-/- mice, all of which were signally reversed by Atorvastatin. Moreover, the percentages of brain Ly6Chi CD45+ cells and CD3+ CD45+ cells, as well as the blood Ly6Chi CD45+ cells, plasma IL-12/IL-23 levels and IL-17 level were found notably increased in ApoE-/- mice, all of which were largely repressed by Atorvastatin. Lastly, the increased BBB permeability, decreased ZO-1 and occludin levels, and reduced KLF2 level were markedly abolished by Atorvastatin. Collectively, Atorvastatin mitigated memory deficits and brain monocyte infiltration in ApoE-/- mice.
Collapse
Affiliation(s)
- Fengchao Gong
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an 271000, Shandong, China
| | - Qian Shi
- Department of Ultrasound, The Second Affiliated Hospital of Shandong First Medical University, Tai'an 271000, Shandong, China
| | - Xiaojie Mou
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an 271000, Shandong, China
| | - Kang Wang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an 271000, Shandong, China
| | - Qianqian Wang
- Department of Ultrasound, The Second Affiliated Hospital of Shandong First Medical University, Tai'an 271000, Shandong, China
| | - Haitao Wang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an 271000, Shandong, China
| |
Collapse
|
10
|
Kennedy CL, Price EM, Mifsud KR, Salatino S, Sharma E, Engledow S, Broxholme J, Goss HM, Reul JM. Genomic regulation of Krüppel-like-factor family members by corticosteroid receptors in the rat brain. Neurobiol Stress 2023; 23:100532. [PMID: 36942087 PMCID: PMC10024234 DOI: 10.1016/j.ynstr.2023.100532] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Hippocampal mineralocorticoid receptors (MRs) and glucocorticoid receptors (GRs) mediate glucocorticoid hormone (GC) action in the hippocampus. These receptors bind to glucocorticoid responsive elements (GREs) within target genes, eliciting transcriptional effects in response to stress and circadian variation. Until recently, little was known about the genome-wide targets of hippocampal MRs and GRs under physiological conditions. Following on from our genome-wide MR and GR ChIP-seq and Ribo-Zero RNA-seq studies on rat hippocampus, we investigated the Krüppel-like factors (KLFs) as targets of MRs and GRs throughout the brain under circadian variation and after acute stress. In particular, Klf2, Klf9 and Klf15 are known to be stress and/or GC responsive and play a role in neurobiological processes including synaptic plasticity and neuronal differentiation. We found increased binding of MR and GR to GREs within Klf2, Klf9 and Klf15 in the hippocampus, amygdala, prefrontal cortex, and neocortex after acute stress and resulting from circadian variation, which was accompanied by upregulation of corresponding hnRNA and mRNA levels. Adrenalectomy abolished transcriptional upregulation of specific Klf genes. These results show that MRs and GRs regulate Klf gene expression throughout the brain following exposure to acute stress or in response to circadian variation, likely alongside other transcription factors.
Collapse
Affiliation(s)
- Clare L.M. Kennedy
- Neuro-Epigenetics Research Group, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Emily M. Price
- Neuro-Epigenetics Research Group, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Karen R. Mifsud
- Neuro-Epigenetics Research Group, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Silvia Salatino
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, United Kingdom
| | - Eshita Sharma
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, United Kingdom
| | - Simon Engledow
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, United Kingdom
| | - John Broxholme
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, United Kingdom
| | - Hannah M. Goss
- Neuro-Epigenetics Research Group, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Johannes M.H.M. Reul
- Neuro-Epigenetics Research Group, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
- Corresponding author.
| |
Collapse
|
11
|
He Z, Du J, Zhang Y, Xu Y, Huang Q, Zhou Q, Wu M, Li Y, Zhang X, Zhang H, Cai Y, Ye K, Wang X, Zhang Y, Han Q, Xiao J. Kruppel-like factor 2 contributes to blood-spinal cord barrier integrity and functional recovery from spinal cord injury by augmenting autophagic flux. Theranostics 2023; 13:849-866. [PMID: 36632224 PMCID: PMC9830435 DOI: 10.7150/thno.74324] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 01/06/2023] Open
Abstract
Background: Increasing evidence suggests that acute traumatic spinal cord injury (SCI)-induced defects in autophagy and autophagy-lysosomal pathway (ALP) may contribute to endothelial barrier disruption following injury. Recently, Kruppel-like factor 2 (KLF2) was reported as a key molecular switch on regulating autophagy. Whether KLF2 coordinates endothelial endothelial ALP in SCI is not known. Methods: Genetic manipulations of KLF2 were performed in bEnd.3 cells and SCI model. Western blot, qRT-PCR, immunofluorescence staining and Lyso-Tracker Red staining, Evans blue dye extravasation, behavioral assessment via Basso mouse scale (BMS), electrophysiology and footprint analysis were performed. Results: In SCI, autophagy flux disruption in endothelial cells contributes to TJ proteins degradation, leading to blood-spinal cord barrier (BSCB) impairment. Furthermore, the KLF2 level was decreased in SCI, overexpression of which alleviated TJ proteins loss and BSCB damage, which improve motor function recovery in SCI mice, while knockdown of KLF2 displayed the opposite effects. At the molecular level, KLF2 overexpression alleviated the TJ proteins degradation and the endothelial permeability by tuning the ALP dysfunction caused by SCI and oxygen glucose deprivation (OGD). Conclusions: Endothelial KLF2 as one of the key contributors to SCI-mediated ALP dysfunction and BSCB disruption. KLF2 could be a promising pharmacological target for the management and treatment of SCI.
Collapse
Affiliation(s)
- Zili He
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, China.,Department of Orthopaedics, Affiliated Pingyang Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Jiqing Du
- Department of Orthopaedics, Affiliated Pingyang Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yu Zhang
- Department of Orthopaedics, Affiliated Pingyang Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yitie Xu
- Department of Orthopaedics, Affiliated Pingyang Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Qian Huang
- Department of Orthopaedics, Affiliated Pingyang Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Qingwei Zhou
- Department of Orthopaedics, Affiliated Pingyang Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Min Wu
- Department of Orthopaedics, Affiliated Pingyang Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yao Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Xie Zhang
- Department of Pharmacy, Ningbo Medical Treatment Center Li Huili Hospital, Ningbo, 315040, China
| | - Hongyu Zhang
- Department of Orthopaedics, Affiliated Pingyang Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yuepiao Cai
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, China.,Department of Orthopaedics, Affiliated Pingyang Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Keyong Ye
- Department of Orthopaedics, Affiliated Pingyang Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiangyang Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Yingze Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, China.,Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051 China
| | - Qi Han
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jian Xiao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, China.,Department of Orthopaedics, Affiliated Pingyang Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| |
Collapse
|
12
|
Wang P, Wang P, Luan H, Wu Y, Chen Y. Midazolam alleviates cellular senescence in SH-SY5Y neuronal cells in Alzheimer's disease. Brain Behav 2023; 13:e2822. [PMID: 36444490 PMCID: PMC9847614 DOI: 10.1002/brb3.2822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/24/2022] [Accepted: 11/01/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) impacts the daily life of aging people. Oligomerized amyloid β (Aβ)-associated neuronal senescence is involved in the pathological mechanism of AD. Blockage of neuronal senescence has been considered an important strategy for the treatment of AD. Midazolam is a hypnotic-sedative drug with pleiotropic properties. AIMS However, the effects of Midazolam in oligomerized Aβ1.42 -induced neurotoxicity have not been reported previously. Here, we investigate whether Midazolam possesses a beneficial effect against oligomerized Aβ1.42 in SH-SY5Y neuronal cells. MATERIALS AND METHODS Cellular senescence was assessed using senescence-associated β-galactosidase staining. Telomerase activity was measured using the TeloTAGGG Telomerase PCR ELISA. RESULTS First, the lactate dehydrogenase release assay demonstrates that 10 and 20 µM are the optimal concentrations of Midazolam used for cell cultures. Senescence-associated β-galactosidase staining results indicate that exposure to oligomerized Aβ1.42 significantly increased cellular senescence of SH-SY5Y cells, but it was significantly alleviated by Midazolam. Additionally, Midazolam restored the oligomerized Aβ1.42 -induced reduction of telomerase activity. Interestingly, we found that oligomerized Aβ1.42 remarkably reduced human telomerase reverse transcriptase (hTERT) gene expression but increased the telomeric repeat-binding factor 2 (TERF2) expression. However, treatment with Midazolam reversed the effects of oligomerized Aβ1.42 on the hTERT and TERF2 gene expressions. Importantly, the presence of Midazolam attenuated Aβ1.42 -induced p53 and p21 expressions. Mechanistically, Midazolam repressed the level of cyclooxygenase-2 (COX-2) and the release of prostaglandin E2. Importantly, overexpression of COX-2 abolished the impact of Midazolam against oligomerized Aβ1.42 in neuronal senescence. CONCLUSION We conclude that the usage of Midazolam is a potential treatment strategy for AD.
Collapse
Affiliation(s)
- Ping Wang
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Xuzhou Medical University Affiliated Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Peipei Wang
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Xuzhou Medical University Affiliated Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Hengfei Luan
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Xuzhou Medical University Affiliated Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Yong Wu
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Xuzhou Medical University Affiliated Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Ying Chen
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Xuzhou Medical University Affiliated Hospital of Lianyungang, Lianyungang, Jiangsu, China
| |
Collapse
|
13
|
Zhou C, Sun P, Hamblin MH, Yin KJ. Genetic deletion of Krüppel-like factor 11 aggravates traumatic brain injury. J Neuroinflammation 2022; 19:281. [PMID: 36403074 PMCID: PMC9675068 DOI: 10.1186/s12974-022-02638-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/06/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The long-term functional recovery of traumatic brain injury (TBI) is hampered by pathological events, such as parenchymal neuroinflammation, neuronal death, and white matter injury. Krüppel-like transcription factor 11 (KLF 11) belongs to the zinc finger family of transcription factors and actively participates in various pathophysiological processes in neurological disorders. Up to now, the role and molecular mechanisms of KLF11 in regulating the pathogenesis of brain trauma is poorly understood. METHODS KLF11 knockout (KO) and wild-type (WT) mice were subjected to experimental TBI, and sensorimotor and cognitive functions were evaluated by rotarod, adhesive tape removal, foot fault, water maze, and passive avoidance tests. Brain tissue loss/neuronal death was examined by MAP2 and NeuN immunostaining, and Cresyl violet staining. White matter injury was assessed by Luxol fast blue staining, and also MBP/SMI32 and Caspr/Nav1.6 immunostaining. Activation of cerebral glial cells and infiltration of blood-borne immune cells were detected by GFAP, Iba-1/CD16/32, Iba-1/CD206, Ly-6B, and F4/80 immunostaining. Brian parenchymal inflammatory cytokines were measured with inflammatory array kits. RESULTS Genetic deletion of KLF11 worsened brain trauma-induced sensorimotor and cognitive deficits, brain tissue loss and neuronal death, and white matter injury in mice. KLF11 genetic deficiency in mice also accelerated post-trauma astrocytic activation, promoted microglial polarization to a pro-inflammatory phenotype, and increased the infiltration of peripheral neutrophils and macrophages into the brain parenchyma. Mechanistically, loss-of-KLF11 function was found to directly increase the expression of pro-inflammatory cytokines in the brains of TBI mice. CONCLUSION KLF11 acts as a novel protective factor in TBI. KLF11 genetic deficiency in mice aggravated the neuroinflammatory responses, grey and white matter injury, and impaired long-term sensorimotor and cognitive recovery. Elucidating the functional importance of KLF11 in TBI may lead us to discover novel pharmacological targets for the development of effective therapies against brain trauma.
Collapse
Affiliation(s)
- Chao Zhou
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15261, USA
- Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh, S514 BST, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - Ping Sun
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15261, USA
- Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh, S514 BST, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - Milton H Hamblin
- Tulane University Health Sciences Center, Tulane University, New Orleans, LA, 70112, USA
- College of Pharmacy, Xavier University of Louisiana, New Orleans, LA, 70125, USA
| | - Ke-Jie Yin
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15261, USA.
- Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh, S514 BST, 200 Lothrop Street, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
14
|
Huang J, Lan H, Xie C, Wei C, Liu Z, Huang Z, Zhou Z, Chen L. Pramipexole Protects Against Traumatic Brain Injury-Induced Blood-Brain Barrier (BBB) Dysfunction. Neurotox Res 2022; 40:1020-1028. [PMID: 35524855 DOI: 10.1007/s12640-022-00495-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/13/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
Abstract
Traumatic brain injury (TBI) is a severe disease of brain damage accompanied by blood-brain barrier (BBB) dysfunction. The BBB is composed of brain microvascular endothelial cells (BMECs), astrocyte terminus, pericytes, and a basement membrane. Tight junction proteins expressed by BMECs play important roles in preserving BBB integrity. Pramipexole is a selective dopamine agonist applied for treating Parkinson's disease and has been recently claimed with neuroprotective capacity. This study will further explore the impact of Pramipexole on tight junctions and BBB integrity to provide the potential treatment strategy for TBI-induced BBB damage. The TBI model was established in mice and was identified by the promoted brain water content, declined Garcia scores, reduced latency of the rotarod test, aggravated pathological changes in the brain cortex, and excessively released inflammatory factors. After treatment with Pramipexole, the neurofunctional deficits, behavioral disability, and aggravated pathological changes were dramatically reversed, accompanied by the alleviated BBB permeability, and upregulated occludin, an important tight junction protein. TBI model cells were established by the scratching bEnd.3 cells method. Cells were stimulated with 10 and 20 μM Pramipexole, followed by exposure to TBI. Increased fluorescence intensity of FITC-dextran, reduced value of TEER, and downregulated occludin and KLF2 were observed in TBI-exposed cells, all of which were greatly reversed by 10 and 20 μM Pramipexole. Furthermore, in KLF2-silenced bEnd.3 cells, the protective ability of Pramipexole against endothelial permeability and the expression level of occludin were dramatically abolished. Collectively, our results suggest that Pramipexole protected against TBI-induced BBB dysfunction by mediating KLF2.
Collapse
Affiliation(s)
- Junping Huang
- Department of Neurosurgery, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530001, Guangxi, China
| | - Huan Lan
- Department of Neurosurgery, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530001, Guangxi, China
| | - Changji Xie
- Department of Neurosurgery, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530001, Guangxi, China
| | - Chengcong Wei
- Department of Neurosurgery, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530001, Guangxi, China
| | - Zhen Liu
- Department of Neurosurgery, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530001, Guangxi, China
| | - Zhixi Huang
- Department of Neurosurgery, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530001, Guangxi, China
| | - Zhiyu Zhou
- Department of Neurosurgery, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530001, Guangxi, China.
| | - Lei Chen
- Department of Neurosurgery, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511500, Guangdong, China.
| |
Collapse
|
15
|
Bu S, Lv Y, Liu Y, Qiao S, Wang H. Zinc Finger Proteins in Neuro-Related Diseases Progression. Front Neurosci 2021; 15:760567. [PMID: 34867169 PMCID: PMC8637543 DOI: 10.3389/fnins.2021.760567] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/11/2021] [Indexed: 01/02/2023] Open
Abstract
Zinc finger proteins (ZNF) are among the most abundant proteins in eukaryotic genomes. It contains several zinc finger domains that can selectively bind to certain DNA or RNA and associate with proteins, therefore, ZNF can regulate gene expression at the transcriptional and translational levels. In terms of neurological diseases, numerous studies have shown that many ZNF are associated with neurological diseases. The purpose of this review is to summarize the types and roles of ZNF in neuropsychiatric disorders. We will describe the structure and classification of ZNF, then focus on the pathophysiological role of ZNF in neuro-related diseases and summarize the mechanism of action of ZNF in neuro-related diseases.
Collapse
Affiliation(s)
- Siyuan Bu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, China
| | - Yihan Lv
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, China
| | - Yusheng Liu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, China
| | - Sen Qiao
- Department of Pharmacology, Center for Molecular Signaling (PZMS), School of Medicine, Saarland University, Homburg, Germany
| | - Hongmei Wang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
16
|
Wang G, Tian F, Li Y, Liu Y, Liu C. Ramelteon Mitigates Free Fatty Acid (FFA)-Induced Attachment of Monocytes to Brain Vascular Endothelial Cells. Neurotox Res 2021; 39:1937-1945. [PMID: 34792763 DOI: 10.1007/s12640-021-00422-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/09/2021] [Accepted: 09/17/2021] [Indexed: 11/25/2022]
Abstract
Acute ischemic stroke is a challenging disease that threatens the life of older people. Dysfunction of brain endothelial cells is reported to be involved in the pathogenesis of acute ischemic stroke. Ramelteon is a novel agonist of melatonin receptor developed for the treatment of insomnia. Recently, the promising protective effect of Ramelteon on brain injury has been widely reported. The present study aims to investigate the protective effect of Ramelteon against free fatty acid (FFA)-induced damages in brain vascular endothelial cells and the underlying mechanism. Firstly, we discovered that Ramelteon administration remarkably reversed the decreased cell viability, increased LDH release, activated oxidative stress, and excessive released inflammatory factors caused by FFAs. Secondly, Ramelteon extensively suppressed the attachment of U937 monocytes to bEnd.3 brain endothelial cells induced by FFAs. In addition, the elevated expression of E-selectin and the reduced expression of KLF2 induced by FFAs were pronouncedly alleviated by Ramelteon. Lastly, silencing of KLF2 abolished the protective effects of Ramelteon against FFA-induced expression of E-selectin and the attachment of U937 monocytes to bEnd.3 brain endothelial cells. In conclusion, Ramelteon mitigated FFA-induced attachment of monocytes to brain vascular endothelial cells by increasing the expression of KLF2 and reducing the expression of E-selectin.
Collapse
Affiliation(s)
- Guijie Wang
- Department of Internal Medicine-Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Fang Tian
- Department of General Medicine, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| | - Yu Li
- Department of Neurosurgery, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| | - Yang Liu
- Department of Internal Medicine-Neurology, the Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, 519041, Guangdong, China
| | - Chunfeng Liu
- Department of Internal Medicine-Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
| |
Collapse
|
17
|
Fraxinellone ameliorates intracerebral hemorrhage-induced secondary brain injury by regulating Krüppel-like transcription factor 2 expression in rats. Brain Res Bull 2021; 177:340-351. [PMID: 34717966 DOI: 10.1016/j.brainresbull.2021.10.018] [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: 04/13/2021] [Revised: 10/11/2021] [Accepted: 10/26/2021] [Indexed: 11/24/2022]
Abstract
Damage to the blood-brain barrier (BBB) is an important factor leading to intracerebral hemorrhage (ICH)-induced secondary brain injury (SBI). Krüppel-like transcription factor 2 (KLF2) plays an important role in the maintenance of the BBB. This study aims to detect the changes of KLF2 after ICH and evaluate the potential effects of fraxinellone on ICH-induced SBI and its correlation with KLF2. An ICH model was established by injecting autologous blood into the right basal ganglia of Sprague-Dawley (SD) rats. First, after ICH induction, the protein levels of KLF2 were reduced. Then, we found that the decrease of KLF2 protein levels induced by ICH could be effectively reversed with the treatment of fraxinellone in vascular endothelial cells. Furthermore, fraxinellone treatment effectively alleviated brain edema, decreased the levels of TNF-α and IL-1β, and improved neuronal cell degeneration induced by ICH. Meanwhile, fraxinellone ameliorated neurobehavioral disorders, motor and sensory impairments, and neurobehavioral disorders and memory loss caused by ICH. Collectively, these findings reveal that KLF2 may be a potential target for fraxinellone to exert neuroprotective effects after ICH, and fraxinellone could be a potential therapeutic agent for SBI after ICH.
Collapse
|
18
|
Han J, Tang H, Yao L, Jin E, Pan W, Chen S. Azilsartan protects against hyperglycemia-induced hyperpermeability of the blood-brain barrier. Bioengineered 2021; 12:3621-3633. [PMID: 34266350 PMCID: PMC8806574 DOI: 10.1080/21655979.2021.1948950] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Diabetes mellitus (DM) is a complex metabolic disease with significant neurological complications and is reported to be closely related to the blood-brain barrier (BBB) disruption. Azilsartan is an antagonist of the Angiotensin II receptor developed for the treatment of hypertension, and it has been recently reported to have neuroprotective effects. The present study aims to investigate the protective effect of Azilsartan against hyperglycemia-induced BBB disruption and its underlying mechanism. Male db/db mice were treated with Azilsartan (20 μg/day) for 10 consecutive days. Compared to the control group, increased BBB permeability, suppressed occludin expression, excessive release of inflammatory factors, and downregulation of krüppel-like factor 2 (KLF2) were observed in diabetic mice, all of which were dramatically reversed by Azilsartan treatment. In the in vitro experiments, elevated endothelial permeability and decreased expression of occludin and KLF2 were observed in high glucose-challenged endothelial cells, which were significantly alleviated by Azilsartan. Lastly, the silencing of KLF2 abolished the protective effects of Azilsartan against the high glucose-induced expression of occludin and endothelial monolayer permeability in bEnd.3 brain endothelial cells. Based on these observations, we concluded that Azilsartan protected against hyperglycemia-induced hyperpermeability of BBB via the KLF2/occludin axis.
Collapse
Affiliation(s)
- Jing Han
- Department of Neurosurgery, the People's Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Hua Tang
- Department of Neurosurgery, the People's Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Longfei Yao
- Department of Neurosurgery, the People's Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Erliang Jin
- Department of Neurosurgery, the People's Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Wanxi Pan
- Department of Neurosurgery, the People's Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Shaojun Chen
- Department of Neurosurgery, the People's Hospital of China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
19
|
Sweet DR, Lam C, Jain MK. Evolutionary Protection of Krüppel-Like Factors 2 and 4 in the Development of the Mature Hemovascular System. Front Cardiovasc Med 2021; 8:645719. [PMID: 34079826 PMCID: PMC8165158 DOI: 10.3389/fcvm.2021.645719] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/21/2021] [Indexed: 02/02/2023] Open
Abstract
A properly functioning hemovascular system, consisting of circulating innate immune cells and endothelial cells (ECs), is essential in the distribution of nutrients to distant tissues while ensuring protection from invading pathogens. Professional phagocytes (e.g., macrophages) and ECs have co-evolved in vertebrates to adapt to increased physiological demands. Intercellular interactions between components of the hemovascular system facilitate numerous functions in physiology and disease in part through the utilization of shared signaling pathways and factors. Krüppel-like factors (KLFs) 2 and 4 are two such transcription factors with critical roles in both cellular compartments. Decreased expression of either factor in myeloid or endothelial cells increases susceptibility to a multitude of inflammatory diseases, underscoring the essential role for their expression in maintaining cellular quiescence. Given the close evolutionary relationship between macrophages and ECs, along with their shared utilization of KLF2 and 4, we hypothesize that KLF genes evolved in such a way that protected their expression in myeloid and endothelial cells. Within this Perspective, we review the roles of KLF2 and 4 in the hemovascular system and explore evolutionary trends in their nucleotide composition that suggest a coordinated protection that corresponds with the development of mature myeloid and endothelial systems.
Collapse
Affiliation(s)
- David R Sweet
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, United States.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States.,Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Cherry Lam
- Department of Biology, New York University, New York, NY, United States
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, United States.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
20
|
Zhao K, Li R, Ruan Q, Meng C, Yin F, Zhu Q. microRNA-125b and its downstream Smurf1/KLF2/ATF2 axis as important promoters on neurological function recovery in rats with spinal cord injury. J Cell Mol Med 2021; 25:5924-5939. [PMID: 33951295 PMCID: PMC8256357 DOI: 10.1111/jcmm.16283] [Citation(s) in RCA: 8] [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: 07/10/2020] [Revised: 12/10/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022] Open
Abstract
The purpose of this study is to investigate the role of microRNA-125b (miR-125b) and its mechanism in spinal cord injury (SCI) by targeting Smurf1. After loss- and gain-function approaches were conducted in SCI rat models and neural stem cells (NSCs) isolated from foetal rats, the Basso-Beattie-Bresnahan (BBB) score was calculated, and related protein expression was determined by Western blot analysis and cell apoptosis by TUNEL staining. NSC viability was detected by CCK-8, migration abilities by Transwell assay and apoptosis by flow cytometry. The relationship between miR-125b, Smurf1 and KLF2 was evaluated by dual-luciferase reporter gene experiments, Co-IP and in vivo ubiquitin modification assays. Inhibition of miR-125b and KLF2 and the up-regulation of Smurf1 and ATF2 were observed in SCI rats. BBB scores were elevated, the expression of Nestin, NeuN, GFAP, NF-200 and Bcl-2 protein was enhanced but that of Bax protein was reduced, and cell apoptosis was inhibited in SCI rats after up-regulating miR-125b or silencing ATF2. Smurf1 was a target gene of miR-125b, which promoted KLF2 degradation through its E3 ubiquitin ligase function, and KLF2 repressed the expression of ATF2 in NSCs. The results in vivo were replicated in vitro. miR-125b overexpression promotes neurological function recovery after SCI.
Collapse
Affiliation(s)
- Kunchi Zhao
- Department of Spine SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Ran Li
- Department of Spine SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Qing Ruan
- Department of Spine SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Chunyang Meng
- Department of Spine SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Fei Yin
- Department of Spine SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Qingsan Zhu
- Department of Spine SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
21
|
Thornton CA, Mulqueen RM, Torkenczy KA, Nishida A, Lowenstein EG, Fields AJ, Steemers FJ, Zhang W, McConnell HL, Woltjer RL, Mishra A, Wright KM, Adey AC. Spatially mapped single-cell chromatin accessibility. Nat Commun 2021; 12:1274. [PMID: 33627658 PMCID: PMC7904839 DOI: 10.1038/s41467-021-21515-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/28/2021] [Indexed: 12/27/2022] Open
Abstract
High-throughput single-cell epigenomic assays can resolve cell type heterogeneity in complex tissues, however, spatial orientation is lost. Here, we present single-cell combinatorial indexing on Microbiopsies Assigned to Positions for the Assay for Transposase Accessible Chromatin, or sciMAP-ATAC, as a method for highly scalable, spatially resolved, single-cell profiling of chromatin states. sciMAP-ATAC produces data of equivalent quality to non-spatial sci-ATAC and retains the positional information of each cell within a 214 micron cubic region, with up to hundreds of tracked positions in a single experiment. We apply sciMAP-ATAC to assess cortical lamination in the adult mouse primary somatosensory cortex and in the human primary visual cortex, where we produce spatial trajectories and integrate our data with non-spatial single-nucleus RNA and other chromatin accessibility single-cell datasets. Finally, we characterize the spatially progressive nature of cerebral ischemic infarction in the mouse brain using a model of transient middle cerebral artery occlusion. Spatial orientation of cells in an interconnected network is lost in high-throughput single-cell epigenomic assays. Here the authors present sciMAP-ATAC to produce spatially resolved single-cell ATAC-seq data.
Collapse
Affiliation(s)
- Casey A Thornton
- Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Ryan M Mulqueen
- Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Kristof A Torkenczy
- Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Andrew Nishida
- Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Eve G Lowenstein
- Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Andrew J Fields
- Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | | | - Wenri Zhang
- Anesthesiology and Peri-Operative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Heather L McConnell
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Randy L Woltjer
- Department of Pathology, Oregon Health & Science University, Portland, OR, USA
| | - Anusha Mishra
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Kevin M Wright
- The Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Andrew C Adey
- Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA. .,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA. .,CEDAR, Oregon Health & Science University, Portland, OR, USA. .,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
22
|
Lu YW, Martino N, Gerlach BD, Lamar JM, Vincent PA, Adam AP, Schwarz JJ. MEF2 (Myocyte Enhancer Factor 2) Is Essential for Endothelial Homeostasis and the Atheroprotective Gene Expression Program. Arterioscler Thromb Vasc Biol 2021; 41:1105-1123. [PMID: 33406884 DOI: 10.1161/atvbaha.120.314978] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Atherosclerosis predominantly forms in regions of oscillatory shear stress while regions of laminar shear stress are protected. This protection is partly through the endothelium in laminar flow regions expressing an anti-inflammatory and antithrombotic gene expression program. Several molecular pathways transmitting these distinct flow patterns to the endothelium have been defined. Our objective is to define the role of the MEF2 (myocyte enhancer factor 2) family of transcription factors in promoting an atheroprotective endothelium. Approach and Results: Here, we show through endothelial-specific deletion of the 3 MEF2 factors in the endothelium, Mef2a, -c, and -d, that MEF2 is a critical regulator of vascular homeostasis. MEF2 deficiency results in systemic inflammation, hemorrhage, thrombocytopenia, leukocytosis, and rapid lethality. Transcriptome analysis reveals that MEF2 is required for normal regulation of 3 pathways implicated in determining the flow responsiveness of the endothelium. Specifically, MEF2 is required for expression of Klf2 and Klf4, 2 partially redundant factors essential for promoting an anti-inflammatory and antithrombotic endothelium. This critical requirement results in phenotypic similarities between endothelial-specific deletions of Mef2a/c/d and Klf2/4. In addition, MEF2 regulates the expression of Notch family genes, Notch1, Dll1, and Jag1, which also promote an atheroprotective endothelium. In contrast to these atheroprotective pathways, MEF2 deficiency upregulates an atherosclerosis promoting pathway through increasing the amount of TAZ (transcriptional coactivator with PDZ-binding motif). CONCLUSIONS Our results implicate MEF2 as a critical upstream regulator of several transcription factors responsible for gene expression programs that affect development of atherosclerosis and promote an anti-inflammatory and antithrombotic endothelium. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Yao Wei Lu
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY
| | - Nina Martino
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY
| | - Brennan D Gerlach
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY
| | - John M Lamar
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY
| | - Peter A Vincent
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY
| | - Alejandro P Adam
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY.,Department of Ophthalmology (A.P.A.), Albany Medical College, NY
| | - John J Schwarz
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY
| |
Collapse
|
23
|
Jin H, Zhu Y, Wang XD, Luo EF, Li YP, Wang BL, Chen YF. BDNF corrects NLRP3 inflammasome-induced pyroptosis and glucose metabolism reprogramming through KLF2/HK1 pathway in vascular endothelial cells. Cell Signal 2020; 78:109843. [PMID: 33253911 DOI: 10.1016/j.cellsig.2020.109843] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022]
Abstract
NLRP3 inflammasome-mediated vascular EC pyroptosis is a key event in the pathogenesis of atherosclerosis. Dysregulation of glucose metabolism is involved in EC dysfunction. Although BDNF plays a protective role in vascular endothelium physiological activity, the mechanisms underlying this activity are not yet clear. In this study, we investigated the role of BDNF in NLRP3 inflammasome-mediated EC pyroptosis and its associated reprogramming of glucose metabolism. HUVECs were treated with human rBDNF under ox-LDL stimulation. rBDNF alleviated ox-LDL-induced NLRP3 inflammasome formation and HUVEC pyroptosis, as evaluated by NLRP3, caspase1-p10, interleukin-18, and interleukin-1β protein levels, co-localization of NLRP3 and apoptosis-associated speck-like protein, and lactate dehydrogenase release. These effects were prevented by tropomyosin receptor kinase B inhibition and KLF2 silencing. The hyper-activation of glycolysis induced by ox-LDL-induced was mitigated by rBDNF via KLF2 as assessed by glucose uptake, lactate production, and extracellular acidification rate. In addition, the BDNF/KLF2 pathway preserved the mitochondrial membrane potential, intracellular reactive oxygen species generation, electron transport chain processing, oxygen consumption rate, and adenosine triphosphate production. Furthermore, KLF2 interacted with HK1 and HK1 overexpression evoked NLRP3 inflammasome formation. At the clinical level, plasma BDNF and lactate levels were measured in 274 patients who underwent computed tomography and coronary angiography for CAD diagnosis. Patients with CAD had lower BDNF and increased lactate levels than those without CAD. In 94 patients with CAD, circulating BDNF levels were inversely associated with lactate levels. In the receiver operating characteristic analysis of CAD, the areas under the curves for 1/BDNF, lactate, and 1/BDNF+lactate were 0.707, 0.702, and 0.753 respectively. These results indicate that BDNF and lactate are linked in atherosclerotic patients, and BDNF inhibits ox-LDL induced NLRP3 inflammasome formation and pyroptosis in HUVECs via KLF2/HK1-mediated glucose metabolism modulation and mitochondrial homeostasis preservation.
Collapse
Affiliation(s)
- Hong Jin
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210000, PR China.
| | - Yi Zhu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210000, PR China
| | - Xiao-Dong Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, PR China
| | - Er-Fei Luo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210000, PR China
| | - Yi-Ping Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210000, PR China
| | - Bi-Lei Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210000, PR China
| | - Yi-Fei Chen
- Department of Cardiology, Xishan Hospital, Wuxi 214000, PR China
| |
Collapse
|
24
|
Fan Y, Wang D, Rao C, Li Y, Rong H, Wang Z, Zhang J. Atorvastatin Combined with Low-Dose Dexamethasone Treatment Protects Endothelial Function Impaired by Chronic Subdural Hematoma via the Transcription Factor KLF-2. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:3291-3299. [PMID: 32848367 PMCID: PMC7429211 DOI: 10.2147/dddt.s256050] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/22/2020] [Indexed: 01/04/2023]
Abstract
Objective Our previous study showed that the combination therapy with atorvastatin and low-dose dexamethasone protected endothelial cell function in chronic subdural hematoma (CSDH) injury. In this study, we aimed to investigate the mechanism underlying the effects of this combination therapy on CSDH-induced cell dysfunction. Methods Monocytes and endothelial cells were cocultured with CSDH patient hematoma samples to mimic the pathological microenvironment of CSDH. Monocytes (THP-1 cells) and endothelial cells (hCMEC/D3 cells) were cocultured in a transwell system for 24 h before stimulation with hematoma samples diluted in endothelial cell medium (ECM) at a 1:1 ratio. Tight junction markers were detected by Western blotting, PCR and immunofluorescence. hCMEC/D3 cells were collected for Western blot and PCR analyses to detect changes in the expression levels of vascular cell adhesion molecule (VCAM-1), intercellular adhesion molecule (ICAM-1), and Kruppel-like factor 2 (KLF-2). The IL-6, IL-10 and VEGF levels in the supernatant were measured by enzyme-linked immunosorbent assay (ELISA). Results KLF-2 expression in endothelial cells was decreased after stimulation with CSDH patient hematoma samples, but combination therapy with atorvastatin and low-dose dexamethasone reversed this trend. KLF-2 protected injured cells by increasing the expression of VE-cadherin and ZO-1; attenuating the expression of VCAM-1, ICAM-1, IL-6 and VEGF; and enhancing the expression of IL-10, all of which play pivotal roles in endothelial inflammation. Moreover, the effect of combination therapy with atorvastatin and low-dose dexamethasone was obviously reduced in endothelial cells with KLF-2 knockdown compared with normal cells. Conclusion Coculture with hematoma samples decreased KLF-2 expression in human cerebral endothelial cells. Combination therapy with atorvastatin and low-dose dexamethasone counteracted hematoma-induced KLF-2 suppression in human cerebral endothelial cells to attenuate robust endothelial inflammation and permeability. KLF-2 plays an important role in drug therapy for CSDH and may become the key factor in treatment and prognosis.
Collapse
Affiliation(s)
- Yueshan Fan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, People's Republic of China.,Graduate School, Tianjin Medical University, Tianjin, 300000, People's Republic of China
| | - Dong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, People's Republic of China
| | - Chenxu Rao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, People's Republic of China.,Graduate School, Tianjin Medical University, Tianjin, 300000, People's Republic of China
| | - Ying Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, People's Republic of China
| | - Hongtao Rong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, People's Republic of China
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, People's Republic of China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, People's Republic of China
| |
Collapse
|
25
|
Sadhukhan R, Leung JWC, Garg S, Krager KJ, Savenka AV, Basnakian AG, Pathak R. Fractionated radiation suppresses Kruppel-like factor 2 pathway to a greater extent than by single exposure to the same total dose. Sci Rep 2020; 10:7734. [PMID: 32382091 PMCID: PMC7206069 DOI: 10.1038/s41598-020-64672-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 04/15/2020] [Indexed: 01/21/2023] Open
Abstract
Kruppel-like factor 2 (KLF2) is a positive transcriptional regulator of several endothelial protective molecules, including thrombomodulin (TM), a surface receptor, and endothelial nitric oxide synthase (eNOS), an enzyme that generates nitric oxide (NO). Loss of TM and eNOS causes endothelial dysfunction, which results in suppressed generation of activated protein C (APC) by TM-thrombin complex and in upregulation of intercellular adhesion molecule 1 (ICAM-1). Mechanistic studies revealed that activation of extracellular signal-regulated kinase 5 (ERK5) via upregulation of myocyte enhancer factor 2 (MEF2) induces KLF2 expression. Radiation causes endothelial dysfunction, but no study has investigated radiation's effects on the KLF2 pathway. Because fractionated radiation is routinely used during cancer radiotherapy, we decided to delineate the effects of radiation dose fractionation on the KLF2 signaling cascade at early time points (up to 24 h). We exposed human primary endothelial cells to radiation as a series of fractionated or as a single exposure, with the same total dose delivered to each group. We measured the expression and activity of critical members of the KLF2 pathway at subsequent time points, and determined whether pharmacological upregulation of KLF2 can reverse the radiation effects. Compared to single exposure, fractionated radiation profoundly suppressed KLF2, TM, and eNOS levels, subdued APC generation, declined KLF2 binding ability to TM and eNOS promoters, enhanced ICAM-1 expression, and decreased expression of upstream regulators of KLF2 (ERK5 and MEF2). Pharmacological inhibitors of the mevalonate pathway prevented fractionated-radiation-induced suppression of KLF2, TM, and eNOS expression. Finally, fractionated irradiation to thoracic region more profoundly suppressed KLF2 and enhanced ICAM-1 expression than single exposure in the lung at 24 h. These data clearly indicate that radiation dose fractionation plays a critical role in modulating levels of KLF2, its upstream regulators, and its downstream target molecules in endothelial cells. Our findings will provide important insights for selecting fractionated regimens during radiotherapy and for developing strategies to alleviate radiotherapy-induced toxicity to healthy tissues.
Collapse
Affiliation(s)
- Ratan Sadhukhan
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Justin W C Leung
- Department of Radiation Oncology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sarthak Garg
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kimberly J Krager
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Alena V Savenka
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Alexei G Basnakian
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Rupak Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
26
|
Kim SM, Ha JS, Han AR, Cho SW, Yang SJ. Effects of α-lipoic acid on LPS-induced neuroinflammation and NLRP3 inflammasome activation through the regulation of BV-2 microglial cells activation. BMB Rep 2020. [PMID: 30940325 PMCID: PMC6827572 DOI: 10.5483/bmbrep.2019.52.10.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Microglial cells are known as the main immune cells in the central nervous system, both regulating its immune response and maintaining its homeostasis. Furthermore, the antioxidant α-lipoic acid (LA) is a recognized therapeutic drug for diabetes because it can easily invade the blood–brain barrier. This study investigated the effect of α-LA on the inflammatory response in lipopolysaccharide (LPS)-treated BV-2 microglial cells. Our results revealed that α-LA significantly attenuated several inflammatory responses in BV-2 microglial cells, including pro-inflammatory cytokines, such as tumor necrosis factor-α and interleukin (IL)-6, and other cytotoxic molecules, such as nitric oxide and reactive oxygen species. In addition, α-LA inhibited the LPS-induced phosphorylation of ERK and p38 and its pharmacological properties were facilitated via the inhibition of the nuclear factor kappa B signaling pathway. Moreover, α-LA suppressed the activation of NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasomes, multiprotein complexes consisting of NLRP3 and caspase-1, which are involved in the innate immune response. Finally, α-LA decreased the genes accountable for the M1 phenotype, IL-1β and ICAM1, whereas it increased the genes responsible for the M2 phenotype, MRC1 and ARG1. These findings suggest that α-LA alleviates the neuroinflammatory response by regulating microglial polarization.
Collapse
Affiliation(s)
- Su Min Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| | - Ji Sun Ha
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| | - A Reum Han
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Seung-Ju Yang
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| |
Collapse
|
27
|
Phoenixin-14 protects human brain vascular endothelial cells against oxygen-glucose deprivation/reoxygenation (OGD/R)-induced inflammation and permeability. Arch Biochem Biophys 2020; 682:108275. [DOI: 10.1016/j.abb.2020.108275] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/08/2020] [Accepted: 01/16/2020] [Indexed: 11/19/2022]
|
28
|
Zhang L, Yuan W, Kong X, Zhang B. Teneligliptin protects against ischemia/reperfusion-induced endothelial permeability in vivo and in vitro. RSC Adv 2020; 10:3765-3774. [PMID: 35492650 PMCID: PMC9048428 DOI: 10.1039/c9ra08810e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/09/2020] [Indexed: 12/13/2022] Open
Abstract
Ischemic stroke is a leading cause of disability and mortality worldwide, especially among the elderly population. Ischemia and reperfusion cause damage to cells and initiate an acute inflammatory response, which leads to cerebral endothelial dysfunction, increased endothelial permeability, and potentially permanent disability. Teneligliptin is a dipeptidyl peptidase-4 (DPP-4) inhibitor that has been used almost exclusively in the treatment of type 2 diabetes mellitus. However, it is still unknown whether teneligliptin possesses a protective effect in brain endothelial dysfunction in the context of ischemic stroke. In the present work, we demonstrate the potential of teneligliptin treatment to protect against ischemia/reperfusion-induced damage using a series of both in vivo and in vitro experiments. Our key findings are that administration of teneligliptin could reduce brain infarct volume, ameliorate neurological damage, and improve brain permeability by increasing the expression of the tight junction protein occludin in middle cerebral artery occlusion (MCAO) mice models. Importantly, teneligliptin displayed a robust protective effect against oxygen–glucose deprivation/reperfusion (OGD/R)-induced cell death of primary human brain microvascular endothelial cells (HBMVECs) in vitro. Notably, teneligliptin prevented OGD/R-induced increased endothelial monolayer permeability in HBMVECs by increasing the expression of occludin, which was mediated by the ERK5/KLF2 signaling pathway. These findings suggest that teneligliptin might serve as a potential therapeutic agent for the treatment of stroke Ischemic stroke is a leading cause of disability and mortality worldwide, especially among the elderly population.![]()
Collapse
Affiliation(s)
- Lei Zhang
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University No. 48 Fenghao West Road, Lianhu District Xi'an Shaanxi province China +86-29-84277356 +86-29-84277356.,Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University Xi'an China
| | - Weiqiong Yuan
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi China
| | - Xiangli Kong
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University No. 48 Fenghao West Road, Lianhu District Xi'an Shaanxi province China +86-29-84277356 +86-29-84277356.,Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University Xi'an China
| | - Bei Zhang
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University No. 48 Fenghao West Road, Lianhu District Xi'an Shaanxi province China +86-29-84277356 +86-29-84277356.,Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University Xi'an China
| |
Collapse
|
29
|
KLF2 protects BV2 microglial cells against oxygen and glucose deprivation injury by modulating BDNF/TrkB pathway. Gene 2019; 735:144277. [PMID: 31821872 DOI: 10.1016/j.gene.2019.144277] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
Abstract
Cerebral ischemia injury is common in cerebral ischemic disease, and treatment options remain limited. Krueppel-like factor 2 (KLF2) is reported to negatively regulate inflammation in several ischemic diseases. Our study aimed to investigate the effects and underlying mechanism of KLF2 in BV2 microglial cells exposed to oxygen and glucose deprivation (OGD). We first found decreased KLF2 and toll-like receptor 2 (TLR2)/TLR4 in these cells. OGD also led to decrease in cell viability and increase in LDH release, apoptosis, the Bax/Bcl-2 ratio, and caspase3/9 expression, as well as production of inflammatory cytokines (e.g., TNFα, IL-1β and IL-6), reactive oxygen species (ROS), and TLR2/TLR4. To examine KLF2's effects on these OGD effects, we infected BV2 microglial cells with an ad-KLF2 or negative control vector, and we found that KLF2 reversed all of the effects of OGD exposure. Furthermore, KLF2 significantly increased levels of BDNF and TrkB in these cells, but these effects were blocked by K252a, a BDNF/TrkB inhibitor. K252a also decreased cell viability and increased apoptosis, inflammatory factors, ROS production, and TLR2/TLR4 expression in OGD-exposed BV2 cells that were treated with KLF2, were implying that K252a could reverse the effects of KLF2 on these cells. Taken together, our study results indicate that KLF2 may protect BV2 microglial cells against OGD injury by activating the BDNF/TrkB pathway.
Collapse
|
30
|
Lasek-Bal A, Kula D, Urbanek T, Puz P, Szymszal J, Jarzab M, Halczok M, Cyplinska R, Bal W, Łabuz-Roszak B, Cieślik A, Jasnos I, Jarzab B, Ziaja D. The Association of SNPs Located in the CDKN2B-AS1 and LPA Genes With Carotid Artery Stenosis and Atherogenic Stroke. Front Neurol 2019; 10:1170. [PMID: 31824394 PMCID: PMC6883000 DOI: 10.3389/fneur.2019.01170] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/21/2019] [Indexed: 11/13/2022] Open
Abstract
Introduction: The aim of this project was to assess the prevalence of four selected SNPs rs4977574 and rs7857345 (CDKN2B-AS1 gene) and rs3798220 and rs10455872 polymorphisms (the LPA gene) in the subpopulation of patients with symptomatic and asymptomatic carotid stenosis. Material and Methods: This study included 623 individuals (244 patients with symptomatic carotid artery stenosis, 176 patients with asymptomatic carotid artery stenosis and 203 healthy people. All the participants underwent neurological examination, duplex Doppler ultrasound examination and molecular procedures. Results: In the first part of the analysis the assiociation of SNPs with stroke/TIA was investigated. The association was seen in symptomatic vs. control group for two SNPs: rs4977574 and rs7857345 (CDKN2B-AS1 gene); genotype distributions for rs4977574 and rs7857345 showed the statistically significant differences between patients and controls (p = 0.043 and 0.017, respectively). No association was observed for rs3798220 and rs10455872 located in the LPA gene. There were statistically significant differences between asymptomatic patients vs. control group in genotype distribution for the SNPs located in CDKN2B-AS1: rs4977574 and rs7857345 (p = 0.031 and 0.0099, respectively); and for the rs3798220 (LPA gene; p = 0.003); however, statistically significant differences did not occur for the rs10455872 polymorphism located in the LPA gene. In the next part of the evaluation, a comparison between symptomatic and asymptomatic patients was performed. Significant differences in genotype distribution were seen only for the rs3798220 polymorphism located in the LPA gene (p = 0.0015). The analysis of the prevalence of the polymorphisms in the total group (symptomatic and asymptomatic) patients in comparison with the control group showed significant differences for three polymorphisms: rs4977574 and rs7857345 (CDKN2B-AS1 gene; p = 0.015 and 0.0046, respectively) and rs3798220 (LPA gene, p = 0.044). Conclusions: The present research on the carotid artery stenosis patient cohort suggests the significant association between the rs4977574, rs7857345 and rs3798220 polymorphisms and carotid artery stenosis as well as between the rs4977574 and rs7857345 polymorphisms and atherogenic stroke. The rs4977574 and rs7857345 polymorphisms in patients with carotid artery stenosis appear to affect a person's susceptibility to atherogenic brain ischemia. Our results need to be replicated in future studies.
Collapse
Affiliation(s)
- Anetta Lasek-Bal
- Department of Neurology, School of Health Sciences, Medical University of Silesia, Katowice, Poland
| | - Dorota Kula
- Maria Skłodowska-Curie, Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Tomasz Urbanek
- Department of General Surgery, Vascular Surgery, Angiology and Phlebology, Medical University of Silesia, Katowice, Poland
| | - Przemysław Puz
- Department of Neurology, School of Health Sciences, Medical University of Silesia, Katowice, Poland
| | - Jan Szymszal
- Faculty of Technical Sciences, University of Occupational Safety Management in Katowice, Katowice, Poland
| | - Michał Jarzab
- 3rd Department of Radiotherapy and Chemotherapy, Maria Sklodowska-Curie Institute-Oncology Center, Gliwice, Poland
| | - Monika Halczok
- Maria Skłodowska-Curie, Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Renata Cyplinska
- Maria Skłodowska-Curie, Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Wiesław Bal
- Department of Outpatient Chemotherapy, Maria Skłodowska-Curie, Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Beata Łabuz-Roszak
- Department of Basic Medical Sciences, Faculty of Public Health, Medical University of Silesia, Katowice, Poland
| | - Aleksandra Cieślik
- Department of Neurology, School of Health Sciences, Medical University of Silesia, Katowice, Poland
| | - Ilona Jasnos
- Department of Neurology, School of Health Sciences, Medical University of Silesia, Katowice, Poland
| | - Barbara Jarzab
- Maria Skłodowska-Curie, Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Damian Ziaja
- Department of General Surgery, Vascular Surgery, Angiology and Phlebology, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
31
|
Kim SM, Ha JS, Han AR, Cho SW, Yang SJ. Effects of α-lipoic acid on LPS-induced neuroinflammation and NLRP3 inflammasome activation through the regulation of BV-2 microglial cells activation. BMB Rep 2019; 52:613-618. [PMID: 30940325 PMCID: PMC6827572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/07/2019] [Accepted: 02/12/2019] [Indexed: 10/15/2023] Open
Abstract
Microglial cells are known as the main immune cells in the central nervous system, both regulating its immune response and maintaining its homeostasis. Furthermore, the antioxidant α-lipoic acid (LA) is a recognized therapeutic drug for diabetes because it can easily invade the blood-brain barrier. This study investigated the effect of α-LA on the inflammatory response in lipopolysaccharide (LPS)-treated BV-2 microglial cells. Our results revealed that α-LA significantly attenuated several inflammatory responses in BV-2 microglial cells, including pro-inflammatory cytokines, such as tumor necrosis factor-α and interleukin (IL)-6, and other cytotoxic molecules, such as nitric oxide and reactive oxygen species. In addition, α-LA inhibited the LPS-induced phosphorylation of ERK and p38 and its pharmacological properties were facilitated via the inhibition of the nuclear factor kappa B signaling pathway. Moreover, α-LA suppressed the activation of NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasomes, multiprotein complexes consisting of NLRP3 and caspase-1, which are involved in the innate immune response. Finally, α-LA decreased the genes accountable for the M1 phenotype, IL-1β and ICAM1, whereas it increased the genes responsible for the M2 phenotype, MRC1 and ARG1. These findings suggest that α-LA alleviates the neuroinflammatory response by regulating microglial polarization. [BMB Reports 2019; 52(10): 613-618].
Collapse
Affiliation(s)
- Su Min Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365,
Korea
| | - Ji Sun Ha
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365,
Korea
| | - A Reum Han
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505,
Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505,
Korea
| | - Seung-Ju Yang
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365,
Korea
| |
Collapse
|
32
|
Rincel M, Aubert P, Chevalier J, Grohard PA, Basso L, Monchaux de Oliveira C, Helbling JC, Lévy É, Chevalier G, Leboyer M, Eberl G, Layé S, Capuron L, Vergnolle N, Neunlist M, Boudin H, Lepage P, Darnaudéry M. Multi-hit early life adversity affects gut microbiota, brain and behavior in a sex-dependent manner. Brain Behav Immun 2019; 80:179-192. [PMID: 30872090 DOI: 10.1016/j.bbi.2019.03.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 02/22/2019] [Accepted: 03/09/2019] [Indexed: 12/15/2022] Open
Abstract
The accumulation of adverse events in utero and during childhood differentially increases the vulnerability to psychiatric diseases in men and women. Gut microbiota is highly sensitive to the early environment and has been recently hypothesized to affect brain development. However, the impact of early-life adversity on gut microbiota, notably with regards to sex differences, remains to be explored. We examined the effects of multifactorial early-life adversity on behavior and microbiota composition in C3H/HeN mice of both sexes exposed to a combination of maternal immune activation (lipopolysaccharide injection on embryonic day 17, 120 µg/kg, i.p.), maternal separation (3hr per day from postnatal day (PND)2 to PND14) and maternal unpredictable chronic mild stress. At adulthood, offspring exposed to multi-hit early adversity showed sex-specific behavioral phenotypes with males exhibiting deficits in social behavior and females showing increased anxiety in the elevated plus maze and increased compulsive behavior in the marble burying test. Early adversity also differentially regulated gene expression in the medial prefrontal cortex (mPFC) according to sex. Interestingly, several genes such as Arc, Btg2, Fosb, Egr4 or Klf2 were oppositely regulated by early adversity in males versus females. Finally, 16S-based microbiota profiling revealed sex-dependent gut dysbiosis. In males, abundance of taxa belonging to Lachnospiraceae and Porphyromonadaceae families or other unclassified Firmicutes, but also Bacteroides, Lactobacillus and Alloprevotella genera was regulated by early adversity. In females, the effects of early adversity were limited and mainly restricted to Lactobacillus and Mucispirillum genera. Our work reveals marked sex differences in a multifactorial model of early-life adversity, both on emotional behaviors and gut microbiota, suggesting that sex should systematically be considered in preclinical studies both in neurogastroenterology and psychiatric research.
Collapse
Affiliation(s)
- Marion Rincel
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Philippe Aubert
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Julien Chevalier
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Pierre-Antoine Grohard
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Lilian Basso
- Institut de Recherche en Santé Digestive, INSERM UMR1220, INRA UMR1416, ENVT, UPS, Toulouse, France
| | - Camille Monchaux de Oliveira
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Jean Christophe Helbling
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Élodie Lévy
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | | | - Marion Leboyer
- Université Paris-est-Créteil, Laboratoire Psychiatrie translationnelle, INSERM U955, Hôpital Chenevier-Mondor, Créteil, France
| | - Gérard Eberl
- Unité Microenvironnement et Immunité, Institut Pasteur, Paris, France
| | - Sophie Layé
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Lucile Capuron
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Nathalie Vergnolle
- Institut de Recherche en Santé Digestive, INSERM UMR1220, INRA UMR1416, ENVT, UPS, Toulouse, France
| | - Michel Neunlist
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Hélène Boudin
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, Univ. Paris-Saclay, Jouy-en-Josas, France
| | - Muriel Darnaudéry
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France.
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Activated protein C (APC) is a homeostatic coagulation protease with anticoagulant and cytoprotective activities. Focusing on APC's effects in the brain, this review discusses three different scenarios that illustrate how APC functions are intimately affecting the physiology and pathophysiology of the brain. RECENT FINDINGS Cytoprotective APC therapy holds promise for the treatment of ischemic stroke, and a recently completed trial suggested that cytoprotective-selective 3K3A-APC reduced bleeding in ischemic stroke patients. In contrast, APC's anticoagulant activity contributes to brain bleeding as shown by the disproportional upregulation of APC generation in cerebral cavernous malformations lesions in mice. However, too little APC generation also contributes to maladies of the brain, such as in case of cerebral malaria where the binding of infected erythrocytes to the endothelial protein C receptor (EPCR) may interfere with the EPCR-dependent functions of the protein C pathway. Furthermore, discoveries of new activities of APC such as the inhibition of the NLRP3-mediated inflammasome and of new applications of APC therapy such as in Alzheimer's disease and graft-versus-host disease continue to advance our knowledge of this important proteolytic regulatory system. SUMMARY APC's many activities or lack thereof are intimately involved in multiple neuropathologies, providing abundant opportunities for translational research.
Collapse
|
34
|
Zhou L, Sun X, Shi Y, Liu J, Luan G, Yang Y. Cysteinyl leukotriene receptor type 1 antagonist montelukast protects against injury of blood-brain barrier. Inflammopharmacology 2019; 27:933-940. [PMID: 31313075 DOI: 10.1007/s10787-019-00611-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 12/14/2018] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB) is formed by tightly connected cerebrovascular endothelial cells. Injury of human brain endothelial cells can cause disruption of the BBB and severe injury to brain tissue. Signals mediated cysteinyl leukotrienes (cysLTs) and their receptors are involved in a variety of pathological conditions. In the current study, our results show that oxygen glucose-deprivation/reoxygenation (OGD/R) induced the expression of leukotriene receptor type 1 (cysLT1R) in brain endothelial cells. Blockage of cysLT1R by its specific antagonist montelukast suppressed OGD/R-induced altered permeability of the human brain endothelial cell (EC) monolayer. Mechanistically, montelukast treatment reversed OGD/R-induced reduction of the tight junction proteins occludin and zonula occludens-1 (ZO-1). Montelukast also ameliorated OGD/R-induced reduction of inhibitors of matrix metalloproteinases (TIMPs), such as TIMP-1 and TIMP-2. On the other hand, montelukast suppressed the expression and production of matrix metalloproteinases (MMPs) and cytokines including MMP-2, MMP-9, interleukin 1β (IL-1β), tumor necrosis factor-α (TNF-α), and interleukin 6 (IL-6). Using a murine middle cerebral artery occlusion brain injury model, we demonstrated that the administration of montelukast improved the surgery-induced brain injury and protected against disruption of brain endothelial junction proteins such as occludin and ZO-1. Collectively, our data suggest that montelukast might confer protective roles against injury in brain endothelial cells.
Collapse
Affiliation(s)
- Li Zhou
- Department of Neurology, Weifang People's Hospital, No. 5, Guangwen Street, Kuiwen District, Weifang, 261000, Shandong, China
| | - Xiaomin Sun
- Department of Neurology, Weifang People's Hospital, No. 5, Guangwen Street, Kuiwen District, Weifang, 261000, Shandong, China
| | - Yong Shi
- Department of Neurology, Weifang People's Hospital, No. 5, Guangwen Street, Kuiwen District, Weifang, 261000, Shandong, China
| | - Junpeng Liu
- Department of Neurology, Weifang People's Hospital, No. 5, Guangwen Street, Kuiwen District, Weifang, 261000, Shandong, China
| | - Guohui Luan
- Department of Neurology, Weifang People's Hospital, No. 5, Guangwen Street, Kuiwen District, Weifang, 261000, Shandong, China
| | - Yanwen Yang
- Department of Neurology, Weifang People's Hospital, No. 5, Guangwen Street, Kuiwen District, Weifang, 261000, Shandong, China.
| |
Collapse
|
35
|
Uric acid treatment after stroke modulates the Krüppel-like factor 2-VEGF-A axis to protect brain endothelial cell functions: Impact of hypertension. Biochem Pharmacol 2019; 164:115-128. [DOI: 10.1016/j.bcp.2019.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/03/2019] [Indexed: 12/29/2022]
|
36
|
Hou Z, Wang Z, Tao Y, Bai J, Yu B, Shen J, Sun H, Xiao L, Xu Y, Zhou J, Wang Z, Geng D. KLF2 regulates osteoblast differentiation by targeting of Runx2. J Transl Med 2019; 99:271-280. [PMID: 30429507 DOI: 10.1038/s41374-018-0149-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 08/26/2018] [Accepted: 09/14/2018] [Indexed: 01/15/2023] Open
Abstract
Osteoblast differentiation plays a critical role in bone formation and maintaining balance in bone remodeling. Runt-related transcription factor 2 (Runx2) is a central transcription factor regulating osteoblast differentiation and promoting bone mineralization. Until now, the molecular regulatory basis and especially the gene regulatory network of osteogenic differentiation have been unclear. Krüppel-like factor 2 (KLF2) is a zinc finger structure and DNA-binding transcription factor. The current study aimed to investigate the physiological function of KLF2 in osteoblast differentiation. Our results indicate that KLF2 is expressed in pre-osteoblast MC3T3-E1 cells and primary osteoblasts. Interestingly, KLF2 expression is increased in osteoblasts during the osteoblastic differentiation process. Overexpression of KLF2 in MC3T3-E1 cells promoted the expression of the osteoblastic differentiation marker genes Alp, Osx, and Ocn, and stimulated mineralization by increasing Runx2 expression at both the mRNA and protein levels. In contrast, knockdown of KLF2 produced the opposite effects. Importantly, we found that KLF2 could physically interact with Runx2. KLF2 promoted osteoblast differentiation by regulating Runx2 and physically interacting with Runx2. Taken together, the findings of this study identify KLF2 as a novel regulator of osteoblast differentiation. Our findings suggest that KLF2 might be a new therapeutic target for bone disease.
Collapse
Affiliation(s)
- Zhenyang Hou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.,Department of Orthopaedics, Tengzhou Central People's Hospital, Tengzhou, Shandong, 277500, China
| | - Zhen Wang
- Department of Orthopaedics, Suzhou Kowloon Hospital Shanghai Jiao Tong University School of Medicine, Suzhou, Jiangsu, 215006, China
| | - Yunxia Tao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Jiaxiang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Binqing Yu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Jining Shen
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Houyi Sun
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Long Xiao
- Department of Orthopaedics, Zhangjiagang Hospital of Traditional Chinese Medicine, Zhangjiagang, Suzhou, Jiangsu, 215600, China
| | - Yaozeng Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Jun Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Zhirong Wang
- Department of Orthopaedics, Zhangjiagang Hospital of Traditional Chinese Medicine, Zhangjiagang, Suzhou, Jiangsu, 215600, China.
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| |
Collapse
|
37
|
Fan Y, Lu H, Liang W, Hu W, Zhang J, Chen YE. Krüppel-like factors and vascular wall homeostasis. J Mol Cell Biol 2018; 9:352-363. [PMID: 28992202 DOI: 10.1093/jmcb/mjx037] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/22/2017] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases (CVDs) are major causes of death worldwide. Identification of promising targets for prevention and treatment of CVDs is paramount in the cardiovascular field. Numerous transcription factors regulate cellular function through modulation of specific genes and thereby are involved in the physiological and pathophysiological processes of CVDs. Although Krüppel-like factors (KLFs) have a similar protein structure with a conserved zinc finger domain, they possess distinct tissue and cell distribution patterns as well as biological functions. In the vascular system, KLF activities are regulated at both transcriptional and posttranscriptional levels. Growing in vitro, in vivo, and genetic epidemiology studies suggest that specific KLFs play important roles in vascular wall biology, which further affect vascular diseases. KLFs regulate various functional aspects such as cell growth, differentiation, activation, and development through controlling a whole cluster of functionally related genes and modulating various signaling pathways in response to pathological conditions. Therapeutic targeting of selective KLF family members may be desirable to achieve distinct treatment effects in the context of various vascular diseases. Further elucidation of the association of KLFs with human CVDs, their underlying molecular mechanisms, and precise protein structure studies will be essential to define KLFs as promising targets for therapeutic interventions in CVDs.
Collapse
Affiliation(s)
- Yanbo Fan
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Haocheng Lu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Wenying Liang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Wenting Hu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jifeng Zhang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
38
|
Saum K, Campos B, Celdran-Bonafonte D, Nayak L, Sangwung P, Thakar C, Roy-Chaudhury P, Owens AP. Uremic Advanced Glycation End Products and Protein-Bound Solutes Induce Endothelial Dysfunction Through Suppression of Krüppel-Like Factor 2. J Am Heart Assoc 2018; 7:e007566. [PMID: 29301761 PMCID: PMC5778969 DOI: 10.1161/jaha.117.007566] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/22/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Cardiovascular disease is the leading cause of morbidity and mortality in patients with end-stage renal disease. The accumulation of uremic solutes in this patient population is associated with endothelial dysfunction and accelerated cardiovascular disease. In this study, we examined the impact of the uremic milieu on the endothelial transcription factor, Krüppel-like factor 2 (KLF2), a key regulator of endothelial function and activation. METHODS AND RESULTS Using serum from uremic pigs with chronic renal insufficiency, our results show that KLF2 expression is suppressed by the uremic milieu and individual uremic solutes in vitro. Specifically, KLF2 expression is significantly decreased in human umbilical vein endothelial cells after treatment with uremic porcine serum or carboxymethyllysine-modified albumin, an advanced glycation end product (AGE) known to induce endothelial dysfunction. AGE-mediated suppression of KLF2 is dependent on activation of the receptor for AGE, as measured by small interfering RNA knockdown of the receptor for AGE. Furthermore, KLF2 suppression promotes endothelial dysfunction, because adenoviral overexpression of KLF2 inhibits reactive oxygen species production and leukocyte adhesion in human umbilical vein endothelial cells. In addition, the application of hemodynamic shear stress, prolonged serum dialysis, or treatment with the receptor for AGE antagonist azeliragon (TTP488) is sufficient to prevent KLF2 suppression in vitro. To decipher the mechanism by which uremic AGEs suppress KLF2 expression, we assessed the role of the receptor for AGE in activation of nuclear factor-κB signaling, a hallmark of endothelial cell activation. Using a constitutively active form of IκBα, we show that translocation of p65 to the nucleus is necessary for KLF2 suppression after treatment with uremic AGEs. CONCLUSIONS These data identify KLF2 suppression as a consequence of the uremic milieu, which may exacerbate endothelial dysfunction and resultant cardiovascular disease.
Collapse
Affiliation(s)
- Keith Saum
- University of Cincinnati Medical Scientist Training Program, The University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Nephrology and Hypertension, The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Begoña Campos
- Division of Nephrology and Hypertension, The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Diego Celdran-Bonafonte
- Division of Nephrology, University of Arizona College of Medicine and Banner University Medical Centers-Tucson and South and Southern Arizona Veterans Affairs Healthcare System, Tucson, AZ
| | - Lalitha Nayak
- Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Panjamaporn Sangwung
- Department of Physiology and Biophysics, Department of Medicine, Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Charuhas Thakar
- Division of Nephrology and Hypertension, The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Prabir Roy-Chaudhury
- Division of Nephrology and Hypertension, The University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Nephrology, University of Arizona College of Medicine and Banner University Medical Centers-Tucson and South and Southern Arizona Veterans Affairs Healthcare System, Tucson, AZ
| | - A Phillip Owens
- Division of Cardiovascular Health and Disease, The University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
39
|
Huang JH, Yin RX, Li WJ, Huang F, Chen WX, Cao XL, Chen YM. Association of the TTC39B rs581080 SNP and serum lipid levels and the risk of coronary artery disease and ischemic stroke. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:11195-11205. [PMID: 31966470 PMCID: PMC6965879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 10/21/2017] [Indexed: 06/10/2023]
Abstract
The tetratricopeptide repeat domain protein 39B gene (TTC39B) single nucleotide polymorphism (SNP) of rs581080 has been associated with serum high-density lipoprotein cholesterol (HDL-C) levels. However, little is known about such association in the Chinese populations. The present study was performed to assess the association between the TTC39B rs581080 SNP and serum lipid levels and the risk of coronary artery disease (CAD) and ischemic stroke (IS) in the Guangxi Han population. Genotypes of the TTC39B rs581080 SNP in 1741 unrelated subjects (CAD, 578; IS, 537; and healthy controls; 624) were determined by the Snapshot Technology. The genotypic and allelic frequencies of the TTC39B rs581080 SNP were different between the CAD/IS patients and controls (P < 0.01 for all). The CG/GG genotypes and G allele were associated with an increased risk of CAD (P = 0.001 for CG/GG vs. CC, P = 0.003 for G vs. C) and IS (P = 0.002 for CG/GG vs. CC; P = 0.004 for G vs. C). The CG/GG genotypes in the healthy controls, but not in CAD or IS patients, were also associated with a decreased serum HDL-C concentration. These results suggest that the TTC39B rs581080 SNP is associated with the risk of CAD and IS in our study population. It is likely to increase the risk of CAD and IS by reducing serum HDL-C levels.
Collapse
Affiliation(s)
- Jian-Hua Huang
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical UniversityNanning, Guangxi, China
| | - Rui-Xing Yin
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical UniversityNanning, Guangxi, China
| | - Wei-Jun Li
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical UniversityNanning, Guangxi, China
| | - Feng Huang
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical UniversityNanning, Guangxi, China
| | - Wu-Xian Chen
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical UniversityNanning, Guangxi, China
| | - Xiao-Li Cao
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical UniversityNanning, Guangxi, China
| | - Yu-Ming Chen
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical UniversityNanning, Guangxi, China
| |
Collapse
|
40
|
Huang RT, Wu D, Meliton A, Oh MJ, Krause M, Lloyd JA, Nigdelioglu R, Hamanaka RB, Jain MK, Birukova A, Kress JP, Birukov KG, Mutlu GM, Fang Y. Experimental Lung Injury Reduces Krüppel-like Factor 2 to Increase Endothelial Permeability via Regulation of RAPGEF3-Rac1 Signaling. Am J Respir Crit Care Med 2017; 195:639-651. [PMID: 27855271 DOI: 10.1164/rccm.201604-0668oc] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
RATIONALE Acute respiratory distress syndrome (ARDS) is caused by widespread endothelial barrier disruption and uncontrolled cytokine storm. Genome-wide association studies (GWAS) have linked multiple genes to ARDS. Although mechanosensitive transcription factor Krüppel-like factor 2 (KLF2) is a major regulator of endothelial function, its role in regulating pulmonary vascular integrity in lung injury and ARDS-associated GWAS genes remains poorly understood. OBJECTIVES To examine KLF2 expression in multiple animal models of acute lung injury and further elucidate the KLF2-mediated pathways involved in endothelial barrier disruption and cytokine storm in experimental lung injury. METHODS Animal and in vitro models of acute lung injury were used to characterize KLF2 expression and its downstream effects responding to influenza A virus (A/WSN/33 [H1N1]), tumor necrosis factor-α, LPS, mechanical stretch/ventilation, or microvascular flow. KLF2 manipulation, permeability measurements, small GTPase activity, luciferase assays, chromatin immunoprecipitation assays, and network analyses were used to determine the mechanistic roles of KLF2 in regulating endothelial monolayer integrity, ARDS-associated GWAS genes, and lung pathophysiology. MEASUREMENTS AND MAIN RESULTS KLF2 is significantly reduced in several animal models of acute lung injury. Microvascular endothelial KLF2 is significantly induced by capillary flow but reduced by pathologic cyclic stretch and inflammatory stimuli. KLF2 is a novel activator of small GTPase Ras-related C3 botulinum toxin substrate 1 by transcriptionally controlling Rap guanine nucleotide exchange factor 3/exchange factor directly activated by cyclic adenosine monophosphate, which maintains vascular integrity. KLF2 regulates multiple ARDS GWAS genes related to cytokine storm, oxidation, and coagulation in lung microvascular endothelium. KLF2 overexpression ameliorates LPS-induced lung injury in mice. CONCLUSIONS Disruption of endothelial KLF2 results in dysregulation of lung microvascular homeostasis and contributes to lung pathology in ARDS.
Collapse
Affiliation(s)
- Ru-Ting Huang
- 1 Section of Pulmonary/Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - David Wu
- 1 Section of Pulmonary/Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Angelo Meliton
- 1 Section of Pulmonary/Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Myung-Jin Oh
- 1 Section of Pulmonary/Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Matthew Krause
- 1 Section of Pulmonary/Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Joyce A Lloyd
- 2 Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia; and
| | - Recep Nigdelioglu
- 1 Section of Pulmonary/Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Robert B Hamanaka
- 1 Section of Pulmonary/Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Mukesh K Jain
- 3 Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, Ohio
| | - Anna Birukova
- 1 Section of Pulmonary/Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - John P Kress
- 1 Section of Pulmonary/Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Konstantin G Birukov
- 1 Section of Pulmonary/Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Gökhan M Mutlu
- 1 Section of Pulmonary/Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Yun Fang
- 1 Section of Pulmonary/Critical Care, Department of Medicine, The University of Chicago, Chicago, Illinois
| |
Collapse
|
41
|
Kaikkonen MU, Halonen P, Liu OHF, Turunen TA, Pajula J, Moreau P, Selvarajan I, Tuomainen T, Aavik E, Tavi P, Ylä-Herttuala S. Genome-Wide Dynamics of Nascent Noncoding RNA Transcription in Porcine Heart After Myocardial Infarction. ACTA ACUST UNITED AC 2017; 10:CIRCGENETICS.117.001702. [DOI: 10.1161/circgenetics.117.001702] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/25/2017] [Indexed: 11/16/2022]
Abstract
Background—
Microarrays and RNA sequencing are widely used to profile transcriptome remodeling during myocardial ischemia. However, the steady-state RNA analysis lacks in sensitivity to detect all noncoding RNA species and does not provide separation between transcriptional and post-transcriptional regulations. Here, we provide the first comprehensive analysis of nascent RNA profiles of mRNAs, primary micro-RNAs, long noncoding RNAs, and enhancer RNAs in a large animal model of acute infarction.
Methods and Results—
Acute infarction was induced by cardiac catheterization of domestic swine. Nuclei isolated from healthy, border zone, and ischemic regions of the affected heart were subjected to global run-on sequencing. Global run-on sequencing analysis indicated that half of affected genes are regulated at the level of transcriptional pausing. A gradient of induction of inflammatory mediators and repression of peroxisome proliferator-activated receptor signaling and oxidative phosphorylation was detected when moving from healthy toward infarcted area. In addition, we interrogated the transcriptional regulation of primary micro-RNAs and provide evidence that several arrhythmia-related target genes exhibit repression at post-transcriptional level. We identified 450 long noncoding RNAs differently regulated by ischemia, including novel conserved long noncoding RNAs expressed in antisense orientation to myocardial transcription factors GATA-binding protein 4, GATA-binding protein 6, and Krüppel-like factor 6. Finally, characterization of enhancers exhibiting differential expression of enhancer RNAs pointed a central role for Krüppel-like factor, MEF2C, ETS, NFY, ATF, E2F2, and NRF1 transcription factors in determining transcriptional responses to ischemia.
Conclusions—
Global run-on sequencing allowed us to follow the gradient of gene expression occurring in the ischemic heart and identify novel noncoding RNAs regulated by oxygen deprivation. These findings highlight potential new targets for diagnosis and treatment of myocardial ischemia.
Collapse
Affiliation(s)
- Minna U. Kaikkonen
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (M.U.K., P.H., O.H.-F.L., T.T., J.P., P.M., I.S., T.T., E.A., P.T., S.Y.-H.); and Heart Center and Gene Therapy Unit, Kuopio University Hospital, Finland (S.Y.-H.)
| | - Paavo Halonen
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (M.U.K., P.H., O.H.-F.L., T.T., J.P., P.M., I.S., T.T., E.A., P.T., S.Y.-H.); and Heart Center and Gene Therapy Unit, Kuopio University Hospital, Finland (S.Y.-H.)
| | - Oscar Hsin-Fu Liu
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (M.U.K., P.H., O.H.-F.L., T.T., J.P., P.M., I.S., T.T., E.A., P.T., S.Y.-H.); and Heart Center and Gene Therapy Unit, Kuopio University Hospital, Finland (S.Y.-H.)
| | - Tiia A. Turunen
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (M.U.K., P.H., O.H.-F.L., T.T., J.P., P.M., I.S., T.T., E.A., P.T., S.Y.-H.); and Heart Center and Gene Therapy Unit, Kuopio University Hospital, Finland (S.Y.-H.)
| | - Juho Pajula
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (M.U.K., P.H., O.H.-F.L., T.T., J.P., P.M., I.S., T.T., E.A., P.T., S.Y.-H.); and Heart Center and Gene Therapy Unit, Kuopio University Hospital, Finland (S.Y.-H.)
| | - Pierre Moreau
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (M.U.K., P.H., O.H.-F.L., T.T., J.P., P.M., I.S., T.T., E.A., P.T., S.Y.-H.); and Heart Center and Gene Therapy Unit, Kuopio University Hospital, Finland (S.Y.-H.)
| | - Ilakya Selvarajan
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (M.U.K., P.H., O.H.-F.L., T.T., J.P., P.M., I.S., T.T., E.A., P.T., S.Y.-H.); and Heart Center and Gene Therapy Unit, Kuopio University Hospital, Finland (S.Y.-H.)
| | - Tomi Tuomainen
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (M.U.K., P.H., O.H.-F.L., T.T., J.P., P.M., I.S., T.T., E.A., P.T., S.Y.-H.); and Heart Center and Gene Therapy Unit, Kuopio University Hospital, Finland (S.Y.-H.)
| | - Einari Aavik
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (M.U.K., P.H., O.H.-F.L., T.T., J.P., P.M., I.S., T.T., E.A., P.T., S.Y.-H.); and Heart Center and Gene Therapy Unit, Kuopio University Hospital, Finland (S.Y.-H.)
| | - Pasi Tavi
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (M.U.K., P.H., O.H.-F.L., T.T., J.P., P.M., I.S., T.T., E.A., P.T., S.Y.-H.); and Heart Center and Gene Therapy Unit, Kuopio University Hospital, Finland (S.Y.-H.)
| | - Seppo Ylä-Herttuala
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (M.U.K., P.H., O.H.-F.L., T.T., J.P., P.M., I.S., T.T., E.A., P.T., S.Y.-H.); and Heart Center and Gene Therapy Unit, Kuopio University Hospital, Finland (S.Y.-H.)
| |
Collapse
|
42
|
Genetic Deletion of Krüppel-Like Factor 11 Aggravates Ischemic Brain Injury. Mol Neurobiol 2017; 55:2911-2921. [PMID: 28456933 DOI: 10.1007/s12035-017-0556-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/12/2017] [Indexed: 02/02/2023]
Abstract
Krüppel-like factors (KLFs) belong to the zinc finger family of transcription factors, and their function in the CNS is largely unexplored. KLF11 is a member of the KLF family, and we have previously demonstrated that peroxisome proliferator-activated receptor gamma-mediated cerebral protection during ischemic insults needs recruitment of KLF11 as its critical coactivator. Here, we sought to determine the role of KLF11 itself in cerebrovascular function and the pathogenesis of ischemic stroke. Transient middle cerebral artery occlusion (MCAO) was performed in KLF11 knockout and wild-type control mice, and brain infarction was analyzed by TTC staining. BBB integrity was assessed by using Evans Blue and TMR-Dextran extravasation assays. KLF11 KO mice exhibited significantly larger brain infarction and poorer neurological outcomes in response to ischemic insults. Genetic deficiency of KLF11 in mice also significantly aggravated ischemia-induced BBB disruption by increasing cerebrovascular permeability and edema. Mechanistically, KLF11 was found to directly regulate IL-6 in the brains of ischemic mice. These findings suggest that KLF11 acts as a novel protective factor in ischemic stroke. Elucidating the functional importance of KLF11 in ischemia may lead us to discover novel pharmacological targets for the development of effective therapies against ischemic stroke.
Collapse
|
43
|
Neuroprotective Effect of 3-(Naphthalen-2-Yl(Propoxy)Methyl)Azetidine Hydrochloride on Brain Ischaemia/Reperfusion Injury. J Neuroimmune Pharmacol 2017; 12:447-461. [PMID: 28247179 DOI: 10.1007/s11481-017-9733-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/21/2017] [Indexed: 01/27/2023]
|
44
|
Sangwung P, Zhou G, Nayak L, Chan ER, Kumar S, Kang DW, Zhang R, Liao X, Lu Y, Sugi K, Fujioka H, Shi H, Lapping SD, Ghosh CC, Higgins SJ, Parikh SM, Jo H, Jain MK. KLF2 and KLF4 control endothelial identity and vascular integrity. JCI Insight 2017; 2:e91700. [PMID: 28239661 DOI: 10.1172/jci.insight.91700] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Maintenance of vascular integrity in the adult animal is needed for survival, and it is critically dependent on the endothelial lining, which controls barrier function, blood fluidity, and flow dynamics. However, nodal regulators that coordinate endothelial identity and function in the adult animal remain poorly characterized. Here, we show that endothelial KLF2 and KLF4 control a large segment of the endothelial transcriptome, thereby affecting virtually all key endothelial functions. Inducible endothelial-specific deletion of Klf2 and/or Klf4 reveals that a single allele of either gene is sufficient for survival, but absence of both (EC-DKO) results in acute death from myocardial infarction, heart failure, and stroke. EC-DKO animals exhibit profound compromise in vascular integrity and profound dysregulation of the coagulation system. Collectively, these studies establish an absolute requirement for KLF2/4 for maintenance of endothelial and vascular integrity in the adult animal.
Collapse
Affiliation(s)
- Panjamaporn Sangwung
- Cardiovascular Research Institute, Department of Medicine, and.,Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Guangjin Zhou
- Cardiovascular Research Institute, Department of Medicine, and
| | - Lalitha Nayak
- Cardiovascular Research Institute, Department of Medicine, and.,Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - E Ricky Chan
- Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Dong-Won Kang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Rongli Zhang
- Cardiovascular Research Institute, Department of Medicine, and
| | - Xudong Liao
- Cardiovascular Research Institute, Department of Medicine, and
| | - Yuan Lu
- Cardiovascular Research Institute, Department of Medicine, and
| | - Keiki Sugi
- Cardiovascular Research Institute, Department of Medicine, and
| | - Hisashi Fujioka
- Electron Microscopy Core Facility, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Hong Shi
- Cardiovascular Research Institute, Department of Medicine, and
| | | | - Chandra C Ghosh
- Center for Vascular Biology Research and Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah J Higgins
- Center for Vascular Biology Research and Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Samir M Parikh
- Center for Vascular Biology Research and Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA.,Division of Cardiology, Emory University, Atlanta, Georgia, USA
| | - Mukesh K Jain
- Cardiovascular Research Institute, Department of Medicine, and.,Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
45
|
Kim J, Kim SM, Na JM, Hahn HG, Cho SW, Yang SJ. Protective effect of 3-(naphthalen-2-yl(propoxy)methyl)azetidine hydrochloride on hypoxia-induced toxicity by suppressing microglial activation in BV-2 cells. BMB Rep 2017; 49:687-692. [PMID: 27756444 PMCID: PMC5346314 DOI: 10.5483/bmbrep.2016.49.12.169] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Indexed: 11/20/2022] Open
Abstract
We recently reported the anti-inflammatory effects of 3-(naphthalen-2-yl(propoxy)methyl)azetidine hydrochloride (KHG26792) on the ATP-induced activation of the NFAT and MAPK pathways through the P2X7 receptor in microglia. To further investigate the underlying mechanism of KHG26792, we studied its protective effects on hypoxia-induced toxicity in microglia. The administration of KHG26792 significantly reduced the hypoxia-induced expression and activity of caspase-3 in BV-2 microglial cells. KHG26792 also reduced hypoxia-induced inducible nitric oxide synthase protein expression, which correlated with reduced nitric oxide accumulation. In addition, KHG26792 attenuated hypoxia-induced protein nitration, reactive oxygen species production, and NADPH oxidase activity. These effects were accompanied by the suppression of hypoxia-induced protein expression of hypoxia-inducible factor 1-alpha and NADPH oxidase-2. Although the clinical relevance of our findings remains to be determined, these data results suggest that KHG26792 prevents hypoxia-induced toxicity by suppressing microglial activation.
Collapse
Affiliation(s)
- Jiae Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Su-Min Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| | - Jung-Min Na
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Hoh-Gyu Hahn
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul 02456, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Seung-Ju Yang
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| |
Collapse
|
46
|
Shan J, Chen L, Lu K. Protective effects of trans-caryophyllene on maintaining osteoblast function. IUBMB Life 2016; 69:22-29. [PMID: 28026135 DOI: 10.1002/iub.1584] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 10/18/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Jinghua Shan
- Department of Anesthesia; Affiliated Hospital of Weifang Medical University; Weifang Shandong Province China
| | - Lixia Chen
- Department of Anesthesia; Affiliated Hospital of Weifang Medical University; Weifang Shandong Province China
| | - Keliang Lu
- Department of Anesthesia; Affiliated Hospital of Weifang Medical University; Weifang Shandong Province China
| |
Collapse
|
47
|
Powers CJ, Dickerson R, Zhang SW, Rink C, Roy S, Sen CK. Human cerebrospinal fluid microRNA: temporal changes following subarachnoid hemorrhage. Physiol Genomics 2016; 48:361-6. [PMID: 26945012 DOI: 10.1152/physiolgenomics.00052.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 03/01/2016] [Indexed: 02/01/2023] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is a devastating form of hemorrhagic stroke with 30-day mortality between 33 and 45%. Delayed cerebral ischemia (DCI) is the chief cause of morbidity and mortality in patients who survive the initial aSAH. DCI accounts for almost 50% of deaths in patients surviving to treatment of the ruptured aneurysm. The mechanisms for brain injury after aSAH and the brain's response to this injury are not fully understood in humans. MicroRNAs (miRs) are 22- to 25-nucleotide single-stranded RNA molecules that inhibit the expression of specific messenger RNA targets. In this work, miR profiling of human cerebrospinal fluid from eight patients after aSAH was performed daily for 10 days with the goal of identifying changes in miR abundance. Using the nanoString nCounter Expression Assay, we identified two specific clusters of miR that were differentially regulated over time. Quantitative RT-PCR was performed on select miRs from each cluster. The first cluster contained miRs known to be present in blood and decreased in abundance over time. miRs in this group include miR-92a and let-7b. The second cluster contained several poorly characterized miRs that increased in abundance over time. miRs in this group included miR-491. This second cluster of miRs may be released into the CSF by the brain itself as a result of the initial SAH. Temporal changes in the abundance of specific miRs in human CSF after aSAH may provide novel insight into the role of miRs in brain injury and the brain's response.
Collapse
Affiliation(s)
- Ciarán J Powers
- Department of Neurological Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio; and
| | - Ryan Dickerson
- Department of Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio
| | - Stacey W Zhang
- Department of Neurological Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio; and
| | - Cameron Rink
- Department of Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio
| | - Sashwati Roy
- Department of Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio
| | - Chandan K Sen
- Department of Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
48
|
Zhang Y, Zou C, Yang S, Fu J. P120 catenin attenuates the angiotensin II-induced apoptosis of human umbilical vein endothelial cells by suppressing the mitochondrial pathway. Int J Mol Med 2016; 37:623-30. [PMID: 26848040 PMCID: PMC4771121 DOI: 10.3892/ijmm.2016.2476] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
Hypertension Hypertension impairs the morphological and functional integrity of circulation. Previous research has shown that the loss of endothelial cells (ECs) is a common event in many cardiovascular diseases. p120 catenin (p120ctn) plays an important role in the regulation of inflammatory responses in ECs. However, the functional significance of p120ctn in angiotensin II (AngII)-induced apoptosis of human umbilical vein endothelial cells (HUVECs) had not previously received much scholarly attention. In the present study, using western blot analysis and RT-PCR, we found that AngII-induced cell apoptosis was correlated with a significant decrease in p120ctn expression. The effect of AngII on cell viability was measured by CCK-8 assay. Knockdown of p120ctn with small hairpin RNA (shRNA) increased AngII-induced apoptosis of HUVECs, as demonstrated by Annexin V/PI staining and flow cytometric analysis. Knockdown of p120ctn with shRNA also increased cytochrome c release into the cytoplasm, and cleaved caspase-3 and -9 protein expression. These were accompanied by a decrease in the Bcl-2/Bax ratio (Bcl-2 and Bax protein expression were measured by western blot analysis), and in mitochondrial membrane potential, as measured using JC-1. Overexpression of p120ctn with adenovirus produced opposite effects. In the present study, we demonstrated that p120ctn attenuated AngII-induced apoptosis of HUVECs through the mitochondria-dependent pathway, suggesting that p120ctn plays a critical role in protecting ECs against apoptosis during hypertension.
Collapse
Affiliation(s)
- Yan Zhang
- Department of VIP Medical Service, Beijing Hospital, Beijing 100730, P.R. China
| | - Chenshuang Zou
- Editorial Department of Chinese Journal of Neuroimmunology and Neurology, Beijing Hospital, Beijing 100730, P.R. China
| | - Shuwen Yang
- Department of VIP Medical Service, Beijing Hospital, Beijing 100730, P.R. China
| | - Jing Fu
- Department of VIP Medical Service, Beijing Hospital, Beijing 100730, P.R. China
| |
Collapse
|
49
|
Shin JA, Jeong SI, Kim M, Yoon JC, Kim HS, Park EM. Visceral adipose tissue inflammation is associated with age-related brain changes and ischemic brain damage in aged mice. Brain Behav Immun 2015; 50:221-231. [PMID: 26184082 DOI: 10.1016/j.bbi.2015.07.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/26/2015] [Accepted: 07/12/2015] [Indexed: 01/22/2023] Open
Abstract
Visceral adipose tissue is accumulated with aging. An increase in visceral fat accompanied by low-grade inflammation is associated with several adult-onset diseases. However, the effects of visceral adipose tissue inflammation on the normal and ischemic brains of aged are not clearly defined. To examine the role of visceral adipose tissue inflammation, we evaluated inflammatory cytokines in the serum, visceral adipose tissue, and brain as well as blood-brain barrier (BBB) permeability in aged male mice (20 months) underwent sham or visceral fat removal surgery compared with the young mice (2.5 months). Additionally, ischemic brain injury was compared in young and aged mice with sham and visceral fat removal surgery. Interleukin (IL)-1β, IL-6, and tumor necrosis factor-α levels in examined organs were increased in aged mice compared with the young mice, and these levels were reduced in the mice with visceral fat removal. Increased BBB permeability with reduced expression of tight junction proteins in aged sham mice were also decreased in mice with visceral fat removal. After focal ischemic injury, aged mice with visceral fat removal showed a reduction in infarct volumes, BBB permeability, and levels of proinflammatory cytokines in the ischemic brain compared with sham mice, although the neurological outcomes were not significantly improved. In addition, further upregulated visceral adipose tissue inflammation in response to ischemic brain injury was attenuated in mice with visceral fat removal. These results suggest that visceral adipose tissue inflammation is associated with age-related changes in the brain and contributes to the ischemic brain damage in the aged mice. We suggest that visceral adiposity should be considered as a factor affecting brain health and ischemic brain damage in the aged population.
Collapse
Affiliation(s)
- Jin A Shin
- Department of Pharmacology, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, 158-710, Republic of Korea
| | - Sae Im Jeong
- Department of Pharmacology, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, 158-710, Republic of Korea
| | - Minsuk Kim
- Department of Pharmacology, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Joo Chun Yoon
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, 158-710, Republic of Korea; Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Hee-Sun Kim
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, 158-710, Republic of Korea; Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Eun-Mi Park
- Department of Pharmacology, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, 158-710, Republic of Korea.
| |
Collapse
|
50
|
Yin KJ, Hamblin M, Fan Y, Zhang J, Chen YE. Krüpple-like factors in the central nervous system: novel mediators in stroke. Metab Brain Dis 2015; 30:401-10. [PMID: 24338065 PMCID: PMC4113556 DOI: 10.1007/s11011-013-9468-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 12/04/2013] [Indexed: 01/08/2023]
Abstract
Transcription factors play an important role in the pathophysiology of many neurological disorders, including stroke. In the past three decades, an increasing number of transcription factors and their related gene signaling networks have been identified, and have become a research focus in the stroke field. Krüppel-like factors (KLFs) are members of the zinc finger family of transcription factors with diverse regulatory functions in cell growth, differentiation, proliferation, migration, apoptosis, metabolism, and inflammation. KLFs are also abundantly expressed in the brain where they serve as critical regulators of neuronal development and regeneration to maintain normal brain function. Dysregulation of KLFs has been linked to various neurological disorders. Recently, there is emerging evidence that suggests KLFs have an important role in the pathogenesis of stroke and provide endogenous vaso-or neuro-protection in the brain's response to ischemic stimuli. In this review, we summarize the basic knowledge and advancement of these transcriptional mediators in the central nervous system, highlighting the novel roles of KLFs in stroke.
Collapse
Affiliation(s)
- Ke-Jie Yin
- Correspondence addressed to: Ke-Jie Yin, M.D., Ph.D., Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Phone: 734-647-8975, Fax: 734-936-2641, , Y. Eugene Chen, M.D., Ph.D., Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Phone: 734-763-7838, Fax: 734-936-2641,
| | | | | | | | - Y. Eugene Chen
- Correspondence addressed to: Ke-Jie Yin, M.D., Ph.D., Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Phone: 734-647-8975, Fax: 734-936-2641, , Y. Eugene Chen, M.D., Ph.D., Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Phone: 734-763-7838, Fax: 734-936-2641,
| |
Collapse
|