101
|
Wu J, He J, Tian X, Li H, Wen Y, Shao Q, Cheng C, Wang G, Sun X. Upregulation of miRNA-9-5p Promotes Angiogenesis after Traumatic Brain Injury by Inhibiting Ptch-1. Neuroscience 2020; 440:160-174. [PMID: 32502567 DOI: 10.1016/j.neuroscience.2020.05.045] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022]
Abstract
MicroRNA-9-5p (miRNA-9-5p) is an important regulator of angiogenesis in many pathological states. However, the effect of miRNA-9-5p on angiogenesis after traumatic brain injury (TBI) has not been elucidated. In this study, a controlled cortical impact (CCI) model was used to induce TBI in Sprague-Dawley rats, and an oxygen glucose deprivation (OGD) model was used to mimic the pathological state in vitro. Brain microvascular endothelial cells (BMECs) were extracted from immature rats. The results showed that the level of miRNA-9-5p was significantly increased in the traumatic foci after TBI, and the upregulation of miRNA9-5p promoted the recovery of neurological function. Moreover, the upregulation of miRNA-9-5p with miRNA agomir significantly increased the density of the microvascular and neurons around the traumatic foci in rats after TBI. The results of the in vitro experiments confirmed that the upregulation of miRNA-9-5p with a miRNA mimic improved cellular viability and alleviated cellular apoptosis. Dual luciferase reporter assay validated that miRNA-9-5p was a posttranscriptional modulator of Ptch-1. Activation of the Hedgehog pathway by increasing the level of miRNA-9-5p promoted the migration and tube formation of BMECs in vitro. In addition, we found that the upregulation of miRNA-9-5p activated the Hedgehog pathway and increased the phosphorylation of AKT, which promoted the expression of cyclin D1, MMP-9 and VEGF in BMECs. All these results indicate that the upregulation of miRNA-9-5p promotes angiogenesis and improves neurological functional recovery after TBI, mainly by activating the Hedgehog pathway. MiRNA-9-5p may be a potential new therapeutic target for TBI.
Collapse
Affiliation(s)
- Jingchuan Wu
- Department of Neurosurgery, General Hospital of The YangTze River Shipping, Wuhan Brain Hospital, Wuhan, Hubei 430014, China; Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Junchi He
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaocui Tian
- College of Pharmacy, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, District of Yuzhong, Chongqing 400016, China
| | - Hui Li
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yi Wen
- Department of Neurosurgery, General Hospital of The YangTze River Shipping, Wuhan Brain Hospital, Wuhan, Hubei 430014, China
| | - Qiang Shao
- Department of Neurosurgery, General Hospital of The YangTze River Shipping, Wuhan Brain Hospital, Wuhan, Hubei 430014, China
| | - Chongjie Cheng
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Guangyu Wang
- Department of Neurosurgery, Qi lu Children's Hospital of Shandong University, Jinan, Shandong 250022, China.
| | - Xiaochuan Sun
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
102
|
Xiang H, Zhang J, Lin C, Zhang L, Liu B, Ouyang L. Targeting autophagy-related protein kinases for potential therapeutic purpose. Acta Pharm Sin B 2020; 10:569-581. [PMID: 32322463 PMCID: PMC7161711 DOI: 10.1016/j.apsb.2019.10.003] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/06/2019] [Accepted: 09/09/2019] [Indexed: 02/08/2023] Open
Abstract
Autophagy, defined as a scavenging process of protein aggregates and damaged organelles mediated by lysosomes, plays a significant role in the quality control of macromolecules and organelles. Since protein kinases are integral to the autophagy process, it is critically important to understand the role of kinases in autophagic regulation. At present, intervention of autophagic processes by small-molecule modulators targeting specific kinases has becoming a reasonable and prevalent strategy for treating several varieties of human disease, especially cancer. In this review, we describe the role of some autophagy-related kinase targets and kinase-mediated phosphorylation mechanisms in autophagy regulation. We also summarize the small-molecule kinase inhibitors/activators of these targets, highlighting the opportunities of these new therapeutic agents.
Collapse
Key Words
- 4E-BP1, eukaryotic translation initiation factor 4E-binding protein
- AKT1, AKT serine/threonine kinase 1
- AMBRA1, autophagy/beclin-1 regulator 1
- AMPK, AMP-activated protein kinase
- ARF, auxin response factor gene
- ATG, autophagy-related protein
- Autophagy
- Autophagy-related kinase
- CaMKK2, calcium/calmodulin-dependent protein kinase kinase 2
- DAPK, death associated protein kinase
- FIP200, FAK family kinase-interacting protein of 200 kDa
- GAP, GTPase-activating protein
- GO, gene ontology
- GSK3α, glycogen synthase kinase 3 alpha
- HMGB1, high mobility group protein B1
- Human disease therapy
- JNK1, C-Jun N-terminal kinase
- LC3, microtubule-associated protein 1 light chain 3
- LKB1, serine/threonine-protein kinase stk11
- LPS, lipopolysaccharide
- LRRK2, leucine rich repeat kinase 2
- PD, Parkinson's disease
- PI, phosphatidylinositol
- PI3 kinase, phosphoinositide 3-kinase
- PI3P, phosphatidylinositol triphosphate
- PIM2, proviral insertion in murine lymphomas 2
- PINK1, PTEN-induced putative kinase 1
- PIP2, phosphatidylinositol-4,5-bisphosphate
- PKACα, a protein kinase cAMP-activated catalytic subunit alpha
- PKCα, protein kinase C alpha type
- PKD1, polycystin-1
- PPIs, protein–protein interactions
- PROTAC, proteolysis targeting chimeras
- PTMs, post-translational modifications
- Phosphorylation
- Protein kinases
- Rheb, the RAS homolog enriched in brain
- Small-molecule kinase inhibitors/activators
- TAK1, transforming growth factor activated kinase-1
- TFEB, transcription factor EB
- TNBC, triple-negative breast cancer
- TSC1/2, tuberous sclerosis complex proteins 1/2
- ULK complex, ULK1–mATG13–FIP200–ATG101 complex
- ULK1, unc-51-like kinase 1
- UVRAG, ultraviolet resistance-associated gene
- mTOR, mammalian target of rapamycin
- mTORC1, mammalian target of rapamycin complex 1
Collapse
Affiliation(s)
- Honggang Xiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Congcong Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lan Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| |
Collapse
|
103
|
Abstract
Coronary heart disease (CHD) is the most common and serious illness in the world and has been researched for many years. However, there are still no real effective ways to prevent and save patients with this disease. When patients present with myocardial infarction, the most important step is to recover ischemic prefusion, which usually is accomplished by coronary artery bypass surgery, coronary artery intervention (PCI), or coronary artery bypass grafting (CABG). These are invasive procedures, and patients with extensive lesions cannot tolerate surgery. It is, therefore, extremely urgent to search for a noninvasive way to save ischemic myocardium. After suffering from ischemia, cardiac or skeletal muscle can partly recover blood flow through angiogenesis (de novo capillary) induced by hypoxia, arteriogenesis, or collateral growth (opening and remodeling of arterioles) triggered by dramatical increase of fluid shear stress (FSS). Evidence has shown that both of them are regulated by various crossed pathways, such as hypoxia-related pathways, cellular metabolism remodeling, inflammatory cells invasion and infiltration, or hemodynamical changes within the vascular wall, but still they do not find effective target for regulating revascularization at present. 5′-Adenosine monophosphate-activated protein kinase (AMPK), as a kinase, is not only an energy modulator but also a sensor of cellular oxygen-reduction substances, and many researches have suggested that AMPK plays an essential role in revascularization but the mechanism is not completely understood. Usually, AMPK can be activated by ADP or AMP, upstream kinases or other cytokines, and pharmacological agents, and then it phosphorylates key molecules that are involved in energy metabolism, autophagy, anti-inflammation, oxidative stress, and aging process to keep cellular homeostasis and finally keeps cell normal activity and function. This review makes a summary on the subunits, activation and downstream targets of AMPK, the mechanism of revascularization, the effects of AMPK in endothelial cells, angiogenesis, and arteriogenesis along with some prospects.
Collapse
|
104
|
Yu B, Wu Y, Li Z. KLF4/Ch25h axis activated by metformin suppresses EndoMT in human umbilical vein endothelial cells. Biochem Biophys Res Commun 2019; 522:838-844. [PMID: 31801667 DOI: 10.1016/j.bbrc.2019.11.181] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 11/26/2019] [Indexed: 01/29/2023]
Abstract
Metformin, an anti-hyperglycemia drug, protected endothelial cells (ECs) from dysfunction while high glucose (HG) caused endothelial dysfunction. Previously, we found that metformin suppressed endothelial-to-mesenchymal transition (EndoMT), a cellular process that promoted endothelial dysfunction. However, the involved mechanism is still unclear. In this study, we found that metformin increased the expression of krüppel-like factor 4 (KLF4) and cholesterol-25-hydroxylase (Ch25h) while HG decreased the expression of KLF4 and Ch25h. In addition, HG promoted EndoMT indicting by the decrease of endothelial maker genes and increase of mesenchymal maker genes. Furthermore, RNA sequence (RNA-seq) data showed that KLF4 suppressed EndoMT. Moreover, we proved that metformin increased Ch25h expression through not only KLF4 but also epigenetic modification including DNA methylation and active histone modification. Lastly, we proved that Ch25h/25 hydroxycholesterol (25 HC)/Liver X receptor α (LXRα) suppressed EndoMT. Altogether, our study demonstrated that KLF4/Ch25h/axis activated by metformin suppressed EndoMT. Therefore, KLF4/Ch25h/axis may be a new potential therapeutic target for endothelial dysfunction diseases.
Collapse
Affiliation(s)
- Beixin Yu
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Yingying Wu
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Zhao Li
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
105
|
Zheng Z, Zhou Y, Ye L, Lu Q, Zhang K, Zhang J, Xie L, Wu Y, Xu K, Zhang H, Xiao J. Histone deacetylase 6 inhibition restores autophagic flux to promote functional recovery after spinal cord injury. Exp Neurol 2019; 324:113138. [PMID: 31794745 DOI: 10.1016/j.expneurol.2019.113138] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/29/2019] [Accepted: 11/29/2019] [Indexed: 12/25/2022]
Abstract
After spinal cord injury (SCI), the inhibitory molecules derived from scars at the lesion sites and the limited regenerative capacity of neuronal axons pose difficulties for the recovery after SCI. Remodeling of cytoskeleton structures including microtubule assembly and tubulin post-translational modification are widely accepted to play a crucial role in initiation of growth cone and regrowth of injured axon. Although increasing studies have focused on the association between tubulin acetylation and autophagy due to the role of tubulin acetylation in organelles and substances transport, there are no studies exploring the effect of tubulin acetylation on autophagy after spinal cord injury (SCI). Here, we found that histone deacetylase 6 (HDAC6) was significantly up-regulated after SCI, while inhibition of HDAC6 by Tubastatin A induced functional recovery after SCI. In view of enzyme-dependent and -independent mechanisms of HDAC6 to adjust diverse cellular processes, such as autophagy, the ubiquitin proteasome system and post-translational modification of tubulin, we mainly focused on the significance of HDAC6 in axonal regeneration and autophagy after SCI. Western blotting, Co-immunoprecipitation and immunofluorescence staining were conducted to showed that Tubastatin A treatment in nocodazole-treated cells and mice suffering from SCI prompted acetylation and stabilization of microtubules and thus restored transport function, which may contribute to restored autophagic flux and increased axonal length. Whereas inhibition of degradation of autolysosomes by bafilomycin A1 (Baf-A1) reversed functional recovery caused by Tubastatin A, revealing the association between tubulin acetylation and autophagy, which supports HDAC6 inhibition as a potential target for SCI treatment.
Collapse
Affiliation(s)
- Zhilong Zheng
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yajiao Zhou
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Luxia Ye
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qi Lu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kairui Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lin Xie
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanqing Wu
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China
| | - Ke Xu
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China
| | - Hongyu Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
106
|
Ashrafizadeh M, Yaribeygi H, Atkin SL, Sahebkar A. Effects of newly introduced antidiabetic drugs on autophagy. Diabetes Metab Syndr 2019; 13:2445-2449. [PMID: 31405658 DOI: 10.1016/j.dsx.2019.06.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/27/2019] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus is a chronic metabolic disorder that has a complex molecular and cellular pathophysiology, resulting in its dynamic progression and that may show differing responses to therapy. The incidence of diabetes mellitus increases with age and requires additive therapeutic agents for its management. SGLT2i and DPP-4 inhibitors and GLP-1 receptor agonists (GLP-1RA) are newly introduced antidiabetic drugs that work through differing mechanisms; DPP-4 inhibitors maintain the endogenous level of GLP1; GLP-1RA result in pharmacological levels of GLP1, whilst SGLT2i act on the proximal tubules of the kidney. They have shown efficacy in the management of diabetes and in contrast to other antidiabetic drugs, do not inherently cause hypoglycemia in therapeutic doses. Autophagy as a highly conserved mechanism to maintain cell survival and homeostasis by degradation of damaged or aged organelles and components, and recognised to be increasingly important in diabetes. In the present review, we discuss the modulatory effects of these newly introduced antidiabetic drugs on the autophagy process.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | | | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|