1
|
Liu T, Zhang J, Chang F, Sun M, He J, Ai D. Role of endothelial Raptor in abnormal arteriogenesis after lower limb ischaemia in type 2 diabetes. Cardiovasc Res 2024; 120:1218-1234. [PMID: 38722901 DOI: 10.1093/cvr/cvae105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 03/08/2024] [Accepted: 03/17/2024] [Indexed: 09/03/2024] Open
Abstract
AIMS Proper arteriogenesis after tissue ischaemia is necessary to rebuild stable blood circulation; nevertheless, this process is impaired in type 2 diabetes mellitus (T2DM). Raptor is a scaffold protein and a component of mammalian target of rapamycin complex 1 (mTORC1). However, the role of the endothelial Raptor in arteriogenesis under the conditions of T2DM remains unknown. This study investigated the role of endothelial Raptor in ischaemia-induced arteriogenesis during T2DM. METHODS AND RESULTS Although endothelial mTORC1 is hyperactive in T2DM, we observed a marked reduction in the expression of endothelial Raptor in two mouse models and in human vessels. Inducible endothelial-specific Raptor knockout severely exacerbated impaired hindlimb perfusion and arteriogenesis after hindlimb ischaemic injury in 12-week high-fat diet fed mice. Additionally, we found that Raptor deficiency dampened vascular endothelial growth factor receptor 2 (VEGFR2) signalling in endothelial cells (ECs) and inhibited VEGF-induced cell migration and tube formation in a PTP1B-dependent manner. Furthermore, mass spectrometry analysis indicated that Raptor interacts with neuropilin 1 (NRP1), the co-receptor of VEGFR2, and mediates VEGFR2 trafficking by facilitating the interaction between NRP1 and Synectin. Finally, we found that EC-specific overexpression of the Raptor mutant (loss of mTOR binding) reversed impaired hindlimb perfusion and arteriogenesis induced by endothelial Raptor knockout in high-fat diet fed mice. CONCLUSION Collectively, our study demonstrated the crucial role of endothelial Raptor in promoting ischaemia-induced arteriogenesis in T2DM by mediating VEGFR2 signalling. Thus, endothelial Raptor is a novel therapeutic target for promoting arteriogenesis and ameliorating perfusion in T2DM.
Collapse
Affiliation(s)
- Ting Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Qixiangtai Rd 22nd, Tianjin 300070, China
| | - Jiachen Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Qixiangtai Rd 22nd, Tianjin 300070, China
| | - Fangyuan Chang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Qixiangtai Rd 22nd, Tianjin 300070, China
| | - Mengyu Sun
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jinlong He
- Department of Physiology and Pathophysiology, Tianjin Medical University, Qixiangtai Rd 22nd, Tianjin 300070, China
| | - Ding Ai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Qixiangtai Rd 22nd, Tianjin 300070, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Qixiangtai Rd 22nd, Tianjin 300070, China
| |
Collapse
|
2
|
Wang Y, Li F, Mao L, Liu Y, Chen S, Liu J, Huang K, Chen Q, Wu J, Lu L, Zheng Y, Shen W, Ying T, Dai Y, Shen Y. Promoting collateral formation in type 2 diabetes mellitus using ultra-small nanodots with autophagy activation and ROS scavenging. J Nanobiotechnology 2024; 22:85. [PMID: 38429826 PMCID: PMC10908163 DOI: 10.1186/s12951-024-02357-z] [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: 01/03/2024] [Accepted: 02/20/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Impaired collateral formation is a major factor contributing to poor prognosis in type 2 diabetes mellitus (T2DM) patients with atherosclerotic cardiovascular disease. However, the current pharmacological treatments for improving collateral formation remain unsatisfactory. The induction of endothelial autophagy and the elimination of reactive oxygen species (ROS) represent potential therapeutic targets for enhancing endothelial angiogenesis and facilitating collateral formation. This study investigates the potential of molybdenum disulfide nanodots (MoS2 NDs) for enhancing collateral formation and improving prognosis. RESULTS Our study shows that MoS2 NDs significantly enhance collateral formation in ischemic tissues of diabetic mice, improving effective blood resupply. Additionally, MoS2 NDs boost the proliferation, migration, and tube formation of endothelial cells under high glucose/hypoxia conditions in vitro. Mechanistically, the beneficial effects of MoS2 NDs on collateral formation not only depend on their known scavenging properties of ROS (H2O2, •O2-, and •OH) but also primarily involve a molecular pathway, cAMP/PKA-NR4A2, which promotes autophagy and contributes to mitigating damage in diabetic endothelial cells. CONCLUSIONS Overall, this study investigated the specific mechanism by which MoS2 NDs mediated autophagy activation and highlighted the synergy between autophagy activation and antioxidation, thus suggesting that an economic and biocompatible nano-agent with dual therapeutic functions is highly preferable for promoting collateral formation in a diabetic context, thus, highlighting their therapeutic potential.
Collapse
Affiliation(s)
- Yixuan Wang
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200025, China
| | - Feifei Li
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200025, China
| | - Linshuang Mao
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200025, China
| | - Yu Liu
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing, 100191, China
| | - Shuai Chen
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200025, China
| | - Jingmeng Liu
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200025, China
| | - Ke Huang
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200025, China
| | - Qiujing Chen
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200025, China
| | - Jianrong Wu
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Lin Lu
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200025, China
| | - Yuanyi Zheng
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Weifeng Shen
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200025, China
| | - Tao Ying
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Yang Dai
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200025, China.
| | - Ying Shen
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200025, China.
| |
Collapse
|
3
|
Zhao N, Yu X, Zhu X, Song Y, Gao F, Yu B, Qu A. Diabetes Mellitus to Accelerated Atherosclerosis: Shared Cellular and Molecular Mechanisms in Glucose and Lipid Metabolism. J Cardiovasc Transl Res 2024; 17:133-152. [PMID: 38091232 DOI: 10.1007/s12265-023-10470-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/23/2023] [Indexed: 02/28/2024]
Abstract
Diabetes is one of the critical independent risk factors for the progression of cardiovascular disease, and the underlying mechanism regarding this association remains poorly understood. Hence, it is urgent to decipher the fundamental pathophysiology and consequently provide new insights into the identification of innovative therapeutic targets for diabetic atherosclerosis. It is now appreciated that different cell types are heavily involved in the progress of diabetic atherosclerosis, including endothelial cells, macrophages, vascular smooth muscle cells, dependence on altered metabolic pathways, intracellular lipids, and high glucose. Additionally, extensive studies have elucidated that diabetes accelerates the odds of atherosclerosis with the explanation that these two chronic disorders share some common mechanisms, such as endothelial dysfunction and inflammation. In this review, we initially summarize the current research and proposed mechanisms and then highlight the role of these three cell types in diabetes-accelerated atherosclerosis and finally establish the mechanism pinpointing the relationship between diabetes and atherosclerosis.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 You'anmen Outer West 1st Street, Beijing, 100069, China
| | - Xiaoting Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 You'anmen Outer West 1st Street, Beijing, 100069, China
| | - Xinxin Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 You'anmen Outer West 1st Street, Beijing, 100069, China
| | - Yanting Song
- Department of Pathology, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| | - Fei Gao
- Department of Cardiology, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| | - Baoqi Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 You'anmen Outer West 1st Street, Beijing, 100069, China.
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, 100069, China.
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 You'anmen Outer West 1st Street, Beijing, 100069, China.
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, 100069, China.
| |
Collapse
|
4
|
Mason EC, Menon S, Schneider BR, Gaskill CF, Dawson MM, Moore CM, Armstrong LC, Cho O, Richmond BW, Kropski JA, West JD, Geraghty P, Gomperts BN, Ess KC, Gally F, Majka SM. Activation of mTOR signaling in adult lung microvascular progenitor cells accelerates lung aging. J Clin Invest 2023; 133:e171430. [PMID: 37874650 PMCID: PMC10721153 DOI: 10.1172/jci171430] [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: 04/12/2023] [Accepted: 10/20/2023] [Indexed: 10/26/2023] Open
Abstract
Reactivation and dysregulation of the mTOR signaling pathway are a hallmark of aging and chronic lung disease; however, the impact on microvascular progenitor cells (MVPCs), capillary angiostasis, and tissue homeostasis is unknown. While the existence of an adult lung vascular progenitor has long been hypothesized, these studies show that Abcg2 enriches for a population of angiogenic tissue-resident MVPCs present in both adult mouse and human lungs using functional, lineage, and transcriptomic analyses. These studies link human and mouse MVPC-specific mTORC1 activation to decreased stemness, angiogenic potential, and disruption of p53 and Wnt pathways, with consequent loss of alveolar-capillary structure and function. Following mTOR activation, these MVPCs adapt a unique transcriptome signature and emerge as a venous subpopulation in the angiodiverse microvascular endothelial subclusters. Thus, our findings support a significant role for mTOR in the maintenance of MVPC function and microvascular niche homeostasis as well as a cell-based mechanism driving loss of tissue structure underlying lung aging and the development of emphysema.
Collapse
Affiliation(s)
- Emma C. Mason
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Swapna Menon
- Pulmonary Vascular Research Institute Kochi and AnalyzeDat Consulting Services, Kerala, India
| | - Benjamin R. Schneider
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Christa F. Gaskill
- Department of Dermatology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Maggie M. Dawson
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Camille M. Moore
- Department of Immunology and Genomic Medicine, Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laura Craig Armstrong
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Okyong Cho
- Genomics and Microarray Core, University of Colorado Cancer Center, Anschutz Medical Center, Aurora, Colorado, USA
| | - Bradley W. Richmond
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center and Department of Veterans Affairs, Nashville, Tennessee, USA
| | - Jonathan A. Kropski
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center and Department of Veterans Affairs, Nashville, Tennessee, USA
| | - James D. West
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center and Department of Veterans Affairs, Nashville, Tennessee, USA
| | - Patrick Geraghty
- Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, New York, USA
| | - Brigitte N. Gomperts
- Translational Research, UCLA Broad Stem Cell Research Center; Pediatrics Division of Pulmonary Medicine, University of California, Los Angeles, California, USA
| | - Kevin C. Ess
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Fabienne Gally
- Department of Immunology and Genomic Medicine, Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Susan M. Majka
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
5
|
Lai TC, Lee CW, Hsu MH, Chen YC, Lin SR, Lin SW, Lee TL, Lin SY, Hsu SH, Tsai JS, Chen YL. Nanocurcumin Reduces High Glucose and Particulate Matter-Induced Endothelial Inflammation: Mitochondrial Function and Involvement of miR-221/222. Int J Nanomedicine 2023; 18:7379-7402. [PMID: 38084125 PMCID: PMC10710795 DOI: 10.2147/ijn.s433658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Purpose Particulate matter (PM) 2.5, harmful air pollutants, and diabetes are associated with high morbidity and mortality from cardiovascular disease (CVD). However, the molecular mechanisms underlying the combined effects of PM and diabetes on CVD remain unclear. Methods Endothelial cells (ECs) treated with high glucose (HG) and PM mimic hyperglycemia and air pollutant exposure in CVD. Endothelial inflammation was evaluated by Western blot and immunofluorescence of ICAM-1 expression and monocyte adhesion. The mechanisms underlying endothelial inflammation were elucidated through MitoSOX Red analysis, JC-1 staining, MitoTracker analysis, and Western blot analysis of mitochondrial fission-related, autophagy-related, and mitophagy-related proteins. Furthermore. nanocurcumin (NCur) pretreatment was used to test if it has a protective effect. Results ECs under co-exposure to HG and PM increased ICAM-1 expression and monocyte adhesion, whereas NCur pretreatment attenuated these changes and improved endothelial inflammation. PM exposure increased mitochondrial ROS levels, worsened mitochondrial membrane potential, promoted mitochondrial fission, induced mitophagy, and aggravated inflammation in HG-treated ECs, while NCur reversed these changes. Also, HG and PM-induced endothelial inflammation is through the JNK signaling pathway and miR-221/222 specifically targeting ICAM-1 and BNIP3. PM exposure also aggravated mitochondrial ROS levels, mitochondrial fission, mitophagy, and endothelial inflammation in STZ-induced hyperglycemic mice, whereas NCur attenuated these changes. Conclusion This study elucidated the mechanisms underlying HG and PM-induced endothelial inflammation in vitro and in vivo. HG and PM treatment increased mitochondrial ROS, mitochondrial fission, and mitophagy in ECs, whereas NCur reversed these conditions. In addition, miR-221/222 plays a role in the amelioration of endothelial inflammation through targeting Bnip3 and ICAM-1, and NCur pretreatment can modulate miR-221/222 levels. Therefore, NCur may be a promising approach to intervene in diabetes and air pollution-induced CVD.
Collapse
Affiliation(s)
- Tsai-Chun Lai
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, Taichung, Taiwan, Republic of China
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Chiang-Wen Lee
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County, Taiwan, Republic of China
- Department of Nursing, Division of Basic Medical Sciences, and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan, Republic of China
- Research Center for Industry of Human Ecology and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan City, Taiwan, Republic of China
| | - Mei-Hsiang Hsu
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Yu-Chen Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan, Republic of China
| | - Shu-Rung Lin
- Department of Bioscience Technology, College of Science, Chung-Yuan Christian University, Taoyuan, Taiwan, Republic of China
- Center for Nanotechnology and Center for Biomedical Technology, Chung-Yuan Christian University, Taoyuan, Taiwan, Republic of China
| | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Tzu-Lin Lee
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Shin-Yu Lin
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Obstetrics and Gynecology, National Taiwan University College of Medicine, Taipei, Taiwan, Republic of China
| | - Shu-Hao Hsu
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Jaw-Shiun Tsai
- Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan, Republic of China
- Center for Complementary and Integrated Medicine, National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| |
Collapse
|
6
|
Jiang X, Yan Q, He J, Zheng Z, Peng X, Cao X, Zhou F, Nie J, Kang T. Interfering with Dusp2 alleviates high glucose-induced vascular endothelial cell dysfunction by promoting p38 MAPK pathway activation. Exp Cell Res 2023; 430:113720. [PMID: 37479052 DOI: 10.1016/j.yexcr.2023.113720] [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: 07/31/2022] [Revised: 06/20/2023] [Accepted: 07/09/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Hyperglycemia-induced vascular endothelial cell dysfunction is a major factor contributing to diabetic lower extremity ischemia. We intend to investigate the role of Dusp2 in hyperglycemia-induced vascular endothelial cell dysfunction and related mechanisms. METHODS The human umbilical vein endothelial cells (HUVECs) were treated with high glucose (HG) as the cell model. Streptozotocin injection was performed to induce diabetes and femoral artery ligation was to induce hind limb ischemia in mice. The levels of Dusp2, p-p38 MAPK, E2F4, and p38 MAPK were evaluated by Western blot or quantitative real-time PCR. The laser Doppler perfusion imaging was conducted to measure blood flow recovery. The cell counting kit-8, transwell, and tube formation assay were performed to evaluate cell proliferation, migration, and angiogenesis, respectively. CD31 immunohistochemical staining was carried out to detect the capillary density of gastrocnemius. The dual-luciferase reporter gene assay and Chromatin immunoprecipitation assay were executed to explore the interaction between E2F4 and Dusp2. RESULTS Dusp2 was highly expressed in HG-induced HUVECs and diabetic lower extremity ischemia model mice. Interference with Dusp2 promoted cell proliferation, migration, and angiogenesis, as well as alleviated mouse diabetic hindlimb ischemia. Dusp2 knockdown up-regulated p-p38 MAPK levels. We verified the binding between E2F4 and Dusp2. Overexpressing E2F4 suppressed Dusp2 levels and promoted cell proliferation, migration, and angiogenesis, co-overexpression of Dusp2 reversed the results. CONCLUSIONS Overexpressing E2F4 promotes endothelial cell proliferation, migration, and angiogenesis by inhibiting Dusp2 expression and activating p38 MAPK to alleviate vascular endothelial cell dysfunction under HG stimulation.
Collapse
Affiliation(s)
- Xinmiao Jiang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Qiong Yan
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jiaqi He
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Zeqi Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiaoping Peng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiaoyan Cao
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Fangbin Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jungang Nie
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Ting Kang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
7
|
Khapchaev AY, Antonova OA, Kazakova OA, Samsonov MV, Vorotnikov AV, Shirinsky VP. Long-Term Experimental Hyperglycemia Does Not Impair Macrovascular Endothelial Barrier Integrity and Function in vitro. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1126-1138. [PMID: 37758312 DOI: 10.1134/s0006297923080072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 10/03/2023]
Abstract
Hyperglycemia is a hallmark of type 2 diabetes implicated in vascular endothelial dysfunction and cardiovascular complications. Many in vitro studies identified endothelial apoptosis as an early outcome of experimentally modeled hyperglycemia emphasizing cell demise as a significant factor of vascular injury. However, endothelial apoptosis has not been observed in vivo until the late stages of type 2 diabetes. Here, we studied the long-term (up to 4 weeks) effects of high glucose (HG, 30 mM) on human umbilical vein endothelial cells (HUVEC) in vitro. HG did not alter HUVEC monolayer morphology, ROS levels, NO production, and exerted minor effects on the HUVEC apoptosis markers. The barrier responses to various clues were indistinguishable from those by cells cultured in physiological glucose (5 mM). Tackling the key regulators of cytoskeletal contractility and endothelial barrier revealed no differences in the histamine-induced intracellular Ca2+ responses, nor in phosphorylation of myosin regulatory light chain or myosin light chain phosphatase. Altogether, these findings suggest that vascular endothelial cells may well tolerate HG for relatively long exposures and warrant further studies to explore mechanisms involved in vascular damage in advanced type 2 diabetes.
Collapse
Affiliation(s)
- Asker Y Khapchaev
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia.
| | - Olga A Antonova
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Olga A Kazakova
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Mikhail V Samsonov
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Alexander V Vorotnikov
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Vladimir P Shirinsky
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| |
Collapse
|
8
|
Liu H, Wang X, Gao H, Yang C, Xie C. Physiological and pathological characteristics of vascular endothelial injury in diabetes and the regulatory mechanism of autophagy. Front Endocrinol (Lausanne) 2023; 14:1191426. [PMID: 37441493 PMCID: PMC10333703 DOI: 10.3389/fendo.2023.1191426] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
Vascular endothelial injury in diabetes mellitus (DM) is the major cause of vascular disease, which is closely related to the occurrence and development of a series of vascular complications and has a serious negative impact on a patient's health and quality of life. The primary function of normal vascular endothelium is to function as a barrier function. However, in the presence of DM, glucose and lipid metabolism disorders, insulin resistance, inflammatory reactions, oxidative stress, and other factors cause vascular endothelial injury, leading to vascular endothelial lesions from morphology to function. Recently, numerous studies have found that autophagy plays a vital role in regulating the progression of vascular endothelial injury. Therefore, this article compares the morphology and function of normal and diabetic vascular endothelium and focuses on the current regulatory mechanisms and the important role of autophagy in diabetic vascular endothelial injury caused by different signal pathways. We aim to provide some references for future research on the mechanism of vascular endothelial injury in DM, investigate autophagy's protective or injurious effect, and study potential drugs using autophagy as a target.
Collapse
Affiliation(s)
- Hanyu Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xueru Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, China
| | - Chan Yang
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
9
|
Wen N, Li J, Zhang W, Li P, Yin X, Zhang W, Wang H, Tang B. Monitoring the Progression of Early Atherosclerosis Using a Fluorescence Nanoprobe for the Detection and Imaging of Phosphorylation and Glucose Levels. Angew Chem Int Ed Engl 2023; 62:e202302161. [PMID: 37072376 DOI: 10.1002/anie.202302161] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/20/2023]
Abstract
Monitoring the early stage of atherosclerosis (AS) without plaque formation is of great significance. Herein, we developed a metal organic framework (MOF)-based fluorescence nanoprobe to analyze the progression of AS by assessing the levels of protein phosphorylation and glucose in blood and tissue. The probe was prepared by post-modification of the MOF with iodine (I3 - )-rhodamine B (RhB) associate, which realizes the specific recognition of target object through the metal joint ZrIV and I3 - -RhB, respectively. We investigated different stages of target object changes in the early non-plaque stage of AS in blood. It was found that the levels of phosphate and glucose in the blood were higher than those of the normal mice. The results of two-photon images showed that early AS mice had higher levels of protein phosphorylation and glucose than that of the normal mice. The present study provides a suitable fluorescence tool for further revealing the pathogenesis and progression of AS.
Collapse
Affiliation(s)
- Ning Wen
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Jin Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Xia Yin
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Wen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
- Laoshan Laboratory, Qingdao, 266237, P. R. China
| |
Collapse
|
10
|
Khater SI, Almanaa TN, Fattah DMA, Khamis T, Seif MM, Dahran N, Alqahtani LS, Metwally MMM, Mostafa M, Albedair RA, Helal AI, Alosaimi M, Mohamed AAR. Liposome-Encapsulated Berberine Alleviates Liver Injury in Type 2 Diabetes via Promoting AMPK/mTOR-Mediated Autophagy and Reducing ER Stress: Morphometric and Immunohistochemical Scoring. Antioxidants (Basel) 2023; 12:1220. [PMID: 37371950 DOI: 10.3390/antiox12061220] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
In the advanced stages of type 2 diabetes mellitus (T2DM), diabetic liver damage is a common complication that can devastate a patient's quality of life. The present study investigated the ability of liposomal berberine (Lip-BBR) to aid in ameliorating hepatic damage and steatosis, insulin homeostasis, and regulating lipid metabolism in type 2 diabetes (T2DM) and the possible pathways by which it does so. Liver tissue microarchitectures and immunohistochemical staining were applied during the study. The rats were divided into a control non-diabetic group and four diabetic groups, which are the T2DM, T2DM-Lip-BBR (10 mg/kg b.wt), T2DM-Vildagliptin (Vild) (10 mg/kg b.wt), and T2DM-BBR-Vild (10 mg/kg b.wt + Vild (5 mg/kg b.wt) groups. The findings demonstrated that Lip-BBR treatment could restore liver tissue microarchitectures, reduce steatosis and liver function, and regulate lipid metabolism. Moreover, Lip-BBR treatment promoted autophagy via the activation of LC3-II and Bclin-1 proteins and activated the AMPK/mTOR pathway in the liver tissue of T2DM rats. Lip-BBR also activated the GLP-1 expression, which stimulated insulin biosynthesis. It decreased the endoplasmic reticulum stress by limiting the CHOP, JNK expression, oxidative stress, and inflammation. Collectively, Lip-BBR ameliorated diabetic liver injury in a T2DM rat model with its promotion activity of AMPK/mTOR-mediated autophagy and limiting ER stress.
Collapse
Affiliation(s)
- Safaa I Khater
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Doaa M Abdel Fattah
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
- Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Mona M Seif
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Naief Dahran
- Department of Anatomy, Faculty of Medicine, University of Jeddah, Jeddah 80203, Saudi Arabia
| | - Leena S Alqahtani
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah 80203, Saudi Arabia
| | - Mohamed M M Metwally
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Mahmoud Mostafa
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Raghad A Albedair
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Azza I Helal
- Department of Histology and Cell Biology, Faculty of Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Manal Alosaimi
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Amany Abdel-Rahman Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| |
Collapse
|
11
|
Luo Q, Chai X, Xin X, Ouyang W, Deng F. Maternal hyperglycemia inhibits pulmonary vasculogenesis during mouse fetal lung development by promoting GβL Ubiquitination-dependent mammalian target of Rapamycin assembly. Diabetol Metab Syndr 2023; 15:49. [PMID: 36927703 PMCID: PMC10021989 DOI: 10.1186/s13098-022-00974-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/24/2022] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is associated with retarded lung development and poor lung health in offspring. Mammalian target of rapamycin (mTOR) is a key regulator of vasculogenesis and angiogenesis. The aim of this study was to investigate the role mTOR plays in pulmonary vasculogenesis during fetal lung development under maternal hyperglycemia. METHODS First, GDM was induced via streptozotocin injection in pregnant C57BL/6 mice before the radial alveolar count (RAC) in the fetal lungs was assessed using hematoxylin and eosin staining. The angiogenic ability of the cultured primary mouse fetal lung endothelial cells (MFLECs) was then assessed using the tube formation assay technique, while western blot and real-time polymerase chain reaction were performed to determine the expression of mTOR, regulatory-associated protein of mTOR (Raptor), rapamycin-insensitive companion of mTOR (Rictor), stress-activated protein kinase interacting protein 1 (Sin1), G protein beta subunit-like protein (GβL), Akt, tumor necrosis receptor associated factor-2 (TRAF2), and OTU deubiquitinase 7B (OTUD7B) in both the fetal lung tissues and the cultured MFLECs. Immunoprecipitation assays were conducted to evaluate the status of GβL-ubiquitination and the association between GβL and mTOR, Raptor, Rictor, and Sin1 in the cultured MFLECs. RESULTS The GDM fetal lungs exhibited a decreased RAC and reduced expression of von Willebrand factor, CD31, and microvessel density. The high glucose level reduced the tube formation ability in the MFLECs, with the mTOR, p-mTOR, p-Raptor, and TRAF2 expression upregulated and the p-Rictor, p-Sin1, p-Akt, and OTUD7B expression downregulated in both the GDM fetal lungs and the high-glucose-treated MFLECs. Meanwhile, GβL-ubiquitination was upregulated in the high-glucose-treated MFLECs along with an increased GβL/Raptor association and decreased GβL/Rictor and GβL/Sin1 association. Furthermore, TRAF2 knockdown inhibited the high-glucose-induced GβL-ubiquitination and GβL/Raptor association and restored the tube formation ability of the MFLECs. CONCLUSION Maternal hyperglycemia inhibits pulmonary vasculogenesis during fetal lung development by promoting GβL-ubiquitination-dependent mTORC1 assembly.
Collapse
Affiliation(s)
- Qingqing Luo
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Obstetrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xinqun Chai
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Xin
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weixiang Ouyang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Feitao Deng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
12
|
Ma J, Chen J, Wang H, Lu D, Liang K. AhR regulates VEGF expression by promoting STAT1 transcriptional activity, thereby affecting endothelial angiogenesis in acute limb ischemia. Chem Biol Interact 2023; 369:110253. [PMID: 36347318 DOI: 10.1016/j.cbi.2022.110253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/07/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Angiogenesis has great potential in the treatment of acute limb ischemia (ALI). Here, we aimed to investigate the effect and mechanism of Aryl hydrocarbon receptor (AhR) on angiogenesis in ALI. METHODS The ALI mouse model was constructed by femoral artery ligation, and the cell ischemia injury was induced by Hypoxia/serum deprivation. The laser doppler perfusion imaging was executed to detect the limb blood flow velocity. The tube formation assay was performed to evaluate angiogenesis. The cell viability was measured by 3-(45)-dimethylthiahiazo(-z-y1)-35-di-phenytetrazoliumromide. The cell migration was detected by wound healing assay. Hematoxylin-eosin, immunohistochemistry, immunofluorescence, dual-luciferase reporter gene assay, and Chromatin immunoprecipitation assay were conducted. RESULTS In ALI models, AhR expression was increased and translocated from cytoplasm to nucleus. Besides, necrosis and inflammatory infiltration were also increased in gastrocnemius tissues of model mice. In addition, AhR loss (LV-sh-AhR) promoted cell viability, angiogenesis, and migration, and also elevated the levels of vascular endothelial growth factor (VEGF), Tie2, and Ang2 in HUVEC models with Hypoxia/serum deprivation injury. Meanwhile, the interaction between AhR and signal transducer and activator of transcription 1 (STAT1), as well as STAT1 and VEGF, has also been confirmed. Co-transfection of LV-sh-AhR and LV-STAT1 suppressed cell viability, angiogenesis, and migration of injured HUVECs. Furthermore, injection of AAV2/9-shAhR in vivo also promoted angiogenesis, which was consistent with the in vitro experimental results. CONCLUSIONS In ALI models, activated AhR was translocated to the nucleus and down-regulated VEGF expression by promoting the transcriptional activity of STAT1, thereby inhibiting endothelial angiogenesis.
Collapse
Affiliation(s)
- Jinhui Ma
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, China.
| | - Jiangbo Chen
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, China
| | - Heng Wang
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, China
| | - Danghui Lu
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, China
| | - Kai Liang
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, China
| |
Collapse
|
13
|
Vorotnikov AV, Khapchaev AY, Nickashin AV, Shirinsky VP. In Vitro Modeling of Diabetes Impact on Vascular Endothelium: Are Essentials Engaged to Tune Metabolism? Biomedicines 2022; 10:biomedicines10123181. [PMID: 36551937 PMCID: PMC9775148 DOI: 10.3390/biomedicines10123181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Angiopathy is a common complication of diabetes mellitus. Vascular endothelium is among the first targets to experience blood-borne metabolic alterations, such as hyperglycemia and hyperlipidemia, the hallmarks of type 2 diabetes. To explore mechanisms of vascular dysfunction and eventual damage brought by these pathologic conditions and to find ways to protect vasculature in diabetic patients, various research approaches are used including in vitro endothelial cell-based models. We present an analysis of the data available from these models that identifies early endothelial cell apoptosis associated with oxidative stress as the major outcome of mimicking hyperglycemia and hyperlipidemia in vitro. However, the fate of endothelial cells observed in these studies does not closely follow it in vivo where massive endothelial damage occurs mainly in the terminal stages of diabetes and in conjunction with comorbidities. We propose that the discrepancy is likely in missing essentials that should be available to cultured endothelial cells to adjust the metabolic state and withstand the immediate apoptosis. We discuss the role of carnitine, creatine, and AMP-activated protein kinase (AMPK) in suiting the endothelial metabolism for long-term function in diabetic type milieu in vitro. Engagement of these essentials is anticipated to expand diabetes research options when using endothelial cell-based models.
Collapse
|
14
|
ShamsEldeen AM, El-Aal SAA, Aboulhoda BE, AbdAllah H, Gamal SM, Hassan FE, Mehesen MN, Rashed LA, Mostafa A, Sadek NB. Combined Systemic Intake of K-ATP Opener (Nicorandil) and Mesenchymal Stem Cells Preconditioned With Nicorandil Alleviates Pancreatic Insufficiency in a Model of Bilateral Renal Ischemia/Reperfusion Injury. Front Physiol 2022; 13:934597. [PMID: 35812319 PMCID: PMC9260271 DOI: 10.3389/fphys.2022.934597] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
We used nicorandil, a K-ATP channel opener, to study the role of these channels in the amelioration of renal ischemia/reperfusion (I/R)-induced pancreatic injury, and the possible involvement of PI3K/Akt/mTOR signaling pathway. Forty-two male Wistar rats were included in this study, six were sacrificed for extraction of bone marrow mesenchymal stem cells (BM-MSCs) and conducting the in-vitro work, the others were included in vivo study and equally divided into six groups. Group 1 (sham control), but groups 2-6 were subjected to bilateral renal I/R: Group 2 (I/R); Group 3 (I/R-NC), treated with nicorandil; Group 4 (I/R-MSCs), treated with BM-MSCs; Group 5 (I/R-MSCC), treated with nicorandil-preconditioned BM-MSCs; Group 6 (I/R-NC-MSCC), treated with both systemic nicorandil and preconditioned BM-MSCC. Renal injury and subsequent pancreatic damage were detected in the I/R group by a significant increase in serum urea, creatinine, fasting glucose, and pancreatic enzymes. The pancreatic tissues showed a reduction in cellularity and a significant decrease in the expression of the cell survival pathway, PI3K/Akt/mTOR, in the I/R group compared to the control. Preconditioning MSCs with nicorandil significantly enhanced the proliferation assay and decreased their apoptotic markers. Indeed, combined systemic nicorandil and nicorandil-preconditioning maintained survival of MSC in the pancreatic tissue and amelioration of apoptotic markers and pancreatic TNF-α production. Histologically, all treated groups revealed better pancreatic architecture, and increased area % of anti-insulin antibody and CD31, which were all best observed in the NC-MSCC group. Thus, using K-ATP channel opener was efficient to enhance PI3K/Akt/mTOR expression levels (in vivo and in vitro).
Collapse
Affiliation(s)
| | | | - Basma Emad Aboulhoda
- Department of Anatomy and Embryology, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Hend AbdAllah
- Department of Anatomy and Embryology, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Sara Mahmoud Gamal
- Department of Physiology, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Fatma E. Hassan
- Department of Physiology, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Marwa Nagi Mehesen
- Department of Medical Pharmacology, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Laila Ahmed Rashed
- Department of Medical Biochemistry and Molecular Biology, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Abeer Mostafa
- Department of Medical Biochemistry and Molecular Biology, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Nermeen Bakr Sadek
- Department of Physiology, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
15
|
Liu Y, Xiang H, Xiong W, Ouyang J, Liu H, Zhao S, Xiao J, Li J, Shu Z, Wang X, Liu H, Zhang J, Fan J, Li Y, Chen S, Lu H. Glucolipotoxicity induces endothelial cell dysfunction by activating autophagy and inhibiting autophagic flow. Diab Vasc Dis Res 2022; 19:14791641221102513. [PMID: 35549572 PMCID: PMC9125420 DOI: 10.1177/14791641221102513] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES This study aims to determine the role and mechanism of autophagy in endothelial cell dysfunction by glucolipotoxicity. METHODS Human umbilical vein endothelial cells (HUVECs) were treated with high glucose and high palmitic acid. The number of autophagosomes was evaluated by monodansylcadaverine (MDC) staining and transmission electron microscopy (TEM). The expression of autophagy-related proteins (LC3 and P62) was assessed by Western blotting. Capillary tube-like formation was evaluated on Matrigel. Reactive oxygen species (ROS) production was detected by DCFH-DA. Cell apoptosis was measured by Hoechst 33258 staining and flow cytometry. Phosphorylation of AMPK, mTOR, and ULK1 was also analyzed by Western blotting. RESULTS We found that glucolipotoxicity induced autophagy initiation and hindered autophagosomes degradation. Moreover, glucolipotoxicity increased the production of intracellular ROS, decreased the ability of tubular formation, and increased cell apoptosis. However, endothelial cell dysfunction was alleviated by 3-methyladenine, an early-stage autophagy inhibitor. Additionally, glucolipotoxicity promoted the phosphorylation of AMPK and ULK1 and inhibited the phosphorylation of mTOR. CONCLUSIONS Glucolipotoxicity initiates autophagy through the AMPK/mTOR/ULK1 signaling pathway and inhibits autophagic flow, leading to the accumulation of autophagosomes, thereby inducing apoptosis and impairing endothelial cell function.
Collapse
Affiliation(s)
- Yulan Liu
- Health Management Center, Third Xiangya Hospital of Central
South University, Changsha, China
- Department of Cardiology, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Hong Xiang
- Center for Experimental Medicine, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Wenfang Xiong
- Department of Cardiology, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Jie Ouyang
- Department of Cardiology, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Hengdao Liu
- Department of Cardiology, The First Affiliated Hospital of
Zhengzhou University, Zhenzhou, China
| | - Shaoli Zhao
- Department of Cardiology, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Jie Xiao
- Department of Cardiology, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Jialing Li
- Department of Cardiology, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Zhihao Shu
- Department of Cardiology, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Xuewen Wang
- Department of Cardiology, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Huiqin Liu
- Department of Cardiology, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Jing Zhang
- Department of Cardiology, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Jianing Fan
- Department of Cardiology, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Ying Li
- Department of Cardiology, Third Xiangya Hospital of Central
South University, Changsha, China
| | - Shuhua Chen
- Departments of Biochemistry, School
of Life Sciences, Central South
University, Changsha, China
- Shuhua Chen, Department of Biochemistry,
School of Life Sciences, Central South University, 172 Tongzipo Road, Changsha
410013, China.
| | - Hongwei Lu
- Health Management Center, Third Xiangya Hospital of Central
South University, Changsha, China
- Center for Experimental Medicine, Third Xiangya Hospital of Central
South University, Changsha, China
- Hongwei Lu, Center for Experimental
Medicine, Third Xiangya Hospital of Central South University, 138 Tongzipo Road,
Changsha 410013, China.
| |
Collapse
|
16
|
Yang Q, Li C, Chen Q. SS31 Ameliorates Oxidative Stress via the Restoration of Autophagic Flux to Protect Aged Mice From Hind Limb Ischemia. Front Cardiovasc Med 2022; 9:789331. [PMID: 35497980 PMCID: PMC9046554 DOI: 10.3389/fcvm.2022.789331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/28/2022] [Indexed: 11/25/2022] Open
Abstract
Background Oxidative stress and impaired autophagic flux play important roles in the development of peripheral artery disease (PAD). SS31 is considered an important antioxidant peptide and autophagy regulator. We aimed to investigate the role of SS31 in PAD myopathy and its possible mechanism both in vivo and in vitro. Methods A hind limb ischemia (HLI) model was established with old C57BL/6 (14-month-old) mice. Mice in the SS31 group were intraperitoneally injected with SS31 (3 mg/kg) for 4 weeks. We examined skeletal muscle function and histomorphology, autophagy-related protein levels and reactive oxygen species (ROS) content. For the in vitro experiments, after C2C12 myotubes were treated with CoCl2, SS31, and chloroquine (CQ) or rapamycin (RAPA), we measured ROS content, autophagy-related protein levels and antioxidant enzyme expression. Results SS31 treatment effectively enhanced the recovery of skeletal muscle function, alleviated skeletal muscle injury and suppressed mitochondrial ROS production in ischemic limbs. SS31 reduced apoptosis and oxidative stress, and SS31 restored impaired autophagic flux by inhibiting the AKT-mTOR pathway. In vitro studies showed that SS31 restored autophagic flux and improved oxidative stress in C2C12 cells. Moreover, phosphorylated AKT (p-AKT) and phosphorylated mTOR (p-mTOR) levels were reduced. Conclusion These experiments indicated that SS31 can inhibit oxidative stress by restoring autophagic flux to reverse hypoxia-induced injury in vivo and in vitro.
Collapse
|
17
|
AKINCI UYSAL Ç, TEMİZ REŞİTOĞLU M, GÜDEN DS, ŞENOL SP, VEZİR Ö, SUCU N, TUNÇTAN B, MALİK KU, FIRAT S. Inhibition of mTOR protects against skeletal muscle and kidney injury following hindlimb ischemia-reperfusion in rats by regulating MERK1/ERK1/2 activity. CUKUROVA MEDICAL JOURNAL 2022. [DOI: 10.17826/cumj.1021518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
18
|
Sun Z, Tong G, Liu Y, Fan H, He W, Wang B, Xia S, He P. Dual Function of a in vivo Albumin-Labeling Tracer for Assessment of Blood Perfusion and Vascular Permeability in Peripheral Arterial Disease by PET. Front Cardiovasc Med 2022; 9:738076. [PMID: 35211521 PMCID: PMC8860820 DOI: 10.3389/fcvm.2022.738076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background Peripheral arterial disease (PAD) leads to tissue ischemia in the extremities. Enhanced vascular permeability plays a critical role in targeted delivery of drugs for effective therapeutic angiogenesis and resultant blood perfusion recovery. However, optimal tracers for evaluating this process in PAD patients are lacking. At this time, we employed a novel in vivo albumin-labeling tracer of dual function, termed as 18F-NEB, to assess blood perfusion as well as vascular permeability by positron emission tomography (PET). Methods and Results After successful establishment of mouse hindlimb ischemia (HI) model, static PET imaging was performed 15 min and 2 h post injection (p.i.) of 18F-NEB at 1, 3, 5, 7, 10 and 14 days post-surgery respectively. Gradual recovery of blood supply was detected by PET scan 15 min p.i. and collaborated by serial Laser Doppler. In addition, the highest vascular permeability observed by high local uptake of 18F-NEB at 2 h p.i. was consistent with histological examinations. Furthermore, we quantitatively evaluated the effect of vascular endothelial growth factor (VEGF) stimulus on vascular permeability and blood perfusion by PET scan using 18F-NEB probe in HI model, which were also confirmed by immunohistological results. Conclusion The application of 18F-NEB probe alone by PET can successfully achieve dual imaging of blood perfusion as well as vascular permeability at different time points p.i. and monitor their responses to therapy in PAD model. The simple labeling approach and multipurpose feature suggest the great promise of using this imaging probe in theranostic applications for treating ischemic disease.
Collapse
Affiliation(s)
- Zhongchan Sun
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Cardiology, Ganzhou Municipal Hospital, Ganzhou, China
- *Correspondence: Zhongchan Sun
| | - Guang Tong
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Cardiac Surgery, Ganzhou Municipal Hospital, Ganzhou, China
| | - Yuanhui Liu
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hualin Fan
- School of Medicine, Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou, China
| | - Weibin He
- School of Medicine, Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou, China
| | - Bo Wang
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shuang Xia
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Pengcheng He
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou, China
- Pengcheng He
| |
Collapse
|
19
|
Xu J, Shan X, Chen C, Gao Y, Zou D, Wang X, Wang T, Shi Y. Tangshenning Attenuates High Glucose-Induced Podocyte Injury via Restoring Autophagy Activity through Inhibiting mTORC1 Activation. J Diabetes Res 2022; 2022:1610416. [PMID: 35799948 PMCID: PMC9256440 DOI: 10.1155/2022/1610416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 12/18/2021] [Accepted: 12/30/2021] [Indexed: 11/20/2022] Open
Abstract
Diabetic nephropathy (DN) is a microvascular complication of diabetes mellitus (DM) and the most common cause of death in diabetic patients. DN progression is associated with podocyte damage due to reduced autophagy caused by mTORC1 activation. Tangshenning (TSN) has been shown to reduce proteinuria, protect renal function, and reduce podocyte damage. Still, the effect of TSN on the autophagic activity of podocytes remains unclear. Herein, in vitro experiments using a high glucose-induced podocyte injury model were performed. Results showed that TSN treatment enhanced the weakened nephrin expression and autophagic activity of podocytes and inhibited the mTORC1 pathway (p-mTOR, mTOR, p-p70S6K, p70S6K, ULK1, and 4EBP1) under high glucose conditions. Furthermore, the mTORC1 activator (siRNA-TSC2) partially inhibited the above beneficial effects of TSN, suggesting that mTORC1 was the target of TSN to regulate autophagy. In summary, TSN reduces podocyte damage induced by high glucose via inhibiting mTORC1 pathway and downstream targets and restoring podocyte autophagy.
Collapse
Affiliation(s)
- Jiayi Xu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Xiaomeng Shan
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Chunwei Chen
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Yanbin Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Dawei Zou
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Xiaolei Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Tao Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Yimin Shi
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| |
Collapse
|
20
|
Liu M, Lu J, Chen Y, Shi X, Li Y, Yang S, Yu J, Guan S. Sodium Sulfite-Induced Mast Cell Pyroptosis and Degranulation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7755-7764. [PMID: 34191510 DOI: 10.1021/acs.jafc.1c02436] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Sodium sulfite, a common food additive, has been proved to cause allergic reaction. Pyroptosis is an inflammatory form of programmed cell death with plasma membrane lysis. In this study, we found that sodium sulfite triggered pyroptosis, which depended on reactive oxygen species (ROS)/NOD-like receptor protein 3 (NLRP3) in RBL-2H3 mast cells. Sodium sulfite increased the generation of ROS and the expression of NLRP3, caspase-1, gasdermin D N-terminal (GSDMD-N), interleukin-1β (IL-1β), and interleukin-18 (IL-18). The ROS scavenger N-acetyl-L-carnosine (NAC) and the NLRP3 inhibitor MCC950 reversed these effects. Furthermore, using a lactate dehydrogenase kit, propidium iodide staining, scanning electron microscopy, colocalization of GSDMD-N with histamine, and neutral red staining, we found that sodium sulfite notably induced cell membrane rupture. Because β-Hexosaminidase and histamine play a key role in allergic reactions, we detected the release of β-Hexosaminidase and histamine. The data showed that the release of β-Hexosaminidase and histamine induced by sodium sulfite was increased with dose independence, which were inhibited after treatment with NAC or MCC950. Overall, evidence suggested that pyroptosis induced by sodium sulfite may rupture the cell membrane and result in degranulation of mast cells. Our study may provide new insights for the mechanism by which sodium sulfite induces mast cell death and sensitization.
Collapse
Affiliation(s)
- Meitong Liu
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Jing Lu
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Yuelin Chen
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Xiaolei Shi
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - YaZhuo Li
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Shuting Yang
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Jing Yu
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Shuang Guan
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
- Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, People's Republic of China
| |
Collapse
|
21
|
Chen D, Liu Y, Chen J, Lin H, Guo H, Wu Y, Xu Y, Zhou Y, Zhou W, Lu R, Zhou J, Wu J. JAK/STAT pathway promotes the progression of diabetic kidney disease via autophagy in podocytes. Eur J Pharmacol 2021; 902:174121. [PMID: 33901462 DOI: 10.1016/j.ejphar.2021.174121] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/13/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022]
Abstract
Diabetic kidney disease (DKD) is one of the major microvascular complications of diabetes and an important cause of end-stage renal disease. Previous studies have shown that the damage to podocyte autophagy is related to the pathogenesis of DKD, and this damage is closely mediated by the Janus kinase (JAK)/signal transductors and the transcription (STAT) signaling pathway. Here, the underlying molecular mechanism of the JAK/STAT signaling pathway regulating podocyte autophagy was investigated. In the present study, compared to controls, DKD mice showed glomerular hypertrophy, increased kidney weight/weight ratio, and increased urinary protein levels, as well as decreased desmin and synaptopodin expression. Meanwhile, levels of triglyceride, total cholesterol, reduced glutathione, and malondialdehyde were also increased in the serum of DKD mice. Further, a lower number of autophagosomes, reduced expression of MAP1LC3 (LC3) in glomeruli, and increased expression of JAK/STAT pathway-related proteins, namely JAK1, JAK2, STAT1, STAT3, STAT5, and STAT6, were observed in DKD mice. In the in vitro experiments, we observed impaired autophagy, enhanced apoptosis, and activated JAK/STAT pathway in podocytes under high glucose conditions. Studies using ruxolitinib inhibitors have showed that suppression of the JAK/STAT pathway in podocytes subjected to high glucose could increase autophagic flux and autophagy-related protein expression. Taken together, the present study demonstrates that high glucose inhibits autophagy by activating the JAK/STAT pathway in mice and podocytes, thereby preventing the efficient removal of damaged proteins and organelles from the body to prevent apoptosis, and ultimately aggravating the progression of podocyte injury and DKD.
Collapse
Affiliation(s)
- Dandan Chen
- Department of Clinical Pharmacy, The Second Aliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Yaoyu Liu
- Department of Clinical Pharmacy, The Second Aliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Junqi Chen
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Hua Lin
- Department of Clinical Pharmacy, The Second Aliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Huijuan Guo
- Department of Pharmacy, Baoan Women's and Children's Hospital, Jinan University, Shenzhen, Guangdong, PR China
| | - Yifan Wu
- Department of Nephrology, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Yuan Xu
- Department of Nephrology, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Yuan Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Wei Zhou
- Department of Clinical Pharmacy, The Second Aliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Ruirui Lu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Jiuyao Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China.
| | - Junbiao Wu
- Department of Clinical Pharmacy, The Second Aliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China.
| |
Collapse
|
22
|
Meng N, Mu X, Gong Y, Wang YH, Zhang J, Wang MH, Yang FY, Jiang CS, Zhang H. Autophagy Induced by a Novel Triazol Derivative Promotes Angiogenesis Through Decreasing Interferon-Inducible Protein 10 Level in Vascular Endothelial Cells. J Cardiovasc Pharmacol 2021; 78:e136-e146. [PMID: 34009854 DOI: 10.1097/fjc.0000000000001034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 04/02/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Autophagy plays an important role in angiogenesis, whereas the mechanisms of vascular endothelial cell (VEC) autophagy associated with angiogenesis remain unclear. In this study, we identified a novel triazol derivative (JL025) that significantly promoted angiogenesis both in vitro and in vivo. Moreover, JL025 had no effects on cell proliferation but dramatically increased the autophagy level of VEC. The suppression of autophagy inhibited JL025-induced angiogenesis in vitro and in vivo, suggesting that JL025-induced angiogenesis was dependent on the enhanced autophagy. Mechanistic studies indicated that JL025-induced VEC autophagy was related to the Protein Kinase B/mTOR signaling pathway. Meanwhile, JL025 decreased the antiangiogenic chemokine interferon-inducible protein 10 (IP10) protein level in human VECs. Importantly, the suppression of autophagy inhibited JL025-induced decrease of IP10 protein level, indicating that autophagy mediated the degradation of IP10. Taken together, our findings provide new insights into the relationship of VEC autophagy with angiogenesis, and JL025 may have a therapeutic potential in related diseases.
Collapse
Affiliation(s)
- Ning Meng
- Department of Vascuar Surgery, School of Biological Science and Technology, University of Jinan, Jinan, China; and
| | - Xin Mu
- Department of Vascuar Surgery, School of Biological Science and Technology, University of Jinan, Jinan, China; and
| | - Yan Gong
- Department of Vascuar Surgery, School of Biological Science and Technology, University of Jinan, Jinan, China; and
| | - Yan Hong Wang
- Department of Vascuar Surgery, School of Biological Science and Technology, University of Jinan, Jinan, China; and
| | - Juan Zhang
- Department of Vascuar Surgery, School of Biological Science and Technology, University of Jinan, Jinan, China; and
| | - Mao Hua Wang
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Feng Ying Yang
- Department of Vascuar Surgery, School of Biological Science and Technology, University of Jinan, Jinan, China; and
| | - Cheng Shi Jiang
- Department of Vascuar Surgery, School of Biological Science and Technology, University of Jinan, Jinan, China; and
| | - Hua Zhang
- Department of Vascuar Surgery, School of Biological Science and Technology, University of Jinan, Jinan, China; and
| |
Collapse
|
23
|
Chen JX, Huang XY, Wang P, Lin WT, Xu WX, Zeng M. Effects and mechanism of arachidonic acid against TNF-α induced apoptosis of endothelial cells. Clin Hemorheol Microcirc 2021; 77:259-265. [PMID: 33337352 DOI: 10.3233/ch-200946] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This study aimed to investigate the effects of arachidonic acid metabolite epoxyeicosatrienoic acid (EETs) in the apoptosis of endothelial cells induced by tumor necrosis factor-alpha (TNF-α). After human umbilical vein endothelial cells were cultured, TNF-α/ActD, 14, 15-EET, and HMR-1098 were added, respectively, into the culture medium. The apoptosis level of endothelial cells was detected by flow cytometry. After TNF-α/ActD induced endothelial cell apoptosis, flow cytometry staining showed that endothelial cell apoptosis increased significantly, and the apoptotic cells were significantly reduced after the addition of 14, 15-EET. However, the apoptotic cells significantly increased after the addition of HMR-1098. Western Blot results showed that the phosphorylation levels of LC3-II and AMPK were increased after TNF-α/ActD induction, and the increase was noticeable after the addition of 14, 15-EET. However, the phosphorylation levels of LC3-II and AMPK significantly decreased after the addition of HMR-1098. The activity of Caspase-8 and -9 decreased significantly after the addition of 14, 15-EET but increased after the addition of HMR-1098. Arachidonic acid can inhibit TNF-α induced endothelial cell apoptosis by upregulating autophagy.
Collapse
Affiliation(s)
- Ji-Xiong Chen
- Department of Medical Care Center, Hainan Provincial People's Hospital, Haikou, China
| | - Xiao-Yan Huang
- Department of Pediatrics, Hainan Maternal and Child Health Hospital, Haikou, China
| | - Ping Wang
- Department of Medical Care Center, Hainan Provincial People's Hospital, Haikou, China
| | - Wen-Ting Lin
- Department of Medical Care Center, Hainan Provincial People's Hospital, Haikou, China
| | - Wen-Xing Xu
- Department of Medical Care Center, Hainan Provincial People's Hospital, Haikou, China
| | - Min Zeng
- Department of Medical Care Center, Hainan Provincial People's Hospital, Haikou, China
| |
Collapse
|
24
|
Zhu Y, Su Y, Zhang J, Zhang Y, Li Y, Han Y, Dong X, Li W, Li W. Astragaloside IV alleviates liver injury in type 2 diabetes due to promotion of AMPK/mTOR‑mediated autophagy. Mol Med Rep 2021; 23:437. [PMID: 33846768 PMCID: PMC8060804 DOI: 10.3892/mmr.2021.12076] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/16/2021] [Indexed: 12/24/2022] Open
Abstract
Diabetic liver injury is a serious complication of type 2 diabetes mellitus (T2DM), which is often irreversible in the later stage, and affects the quality of life. Autophagy serves an important role in the occurrence and development of diabetic liver injury. For example, it can improve insulin resistance (IR), dyslipidaemia, oxidative stress and inflammation. Astragaloside IV (AS-IV) is a natural saponin isolated from the plant Astragalus membranaceus, which has comprehensive pharmacological effects, such as anti-oxidation, anti-inflammation and anti-apoptosis properties, as well as can enhance immunity. However, whether AS-IV can alleviate diabetic liver injury in T2DM and its underlying mechanisms remain unknown. The present study used high-fat diets combined with low-dose streptozotocin to induce a diabetic liver injury model in T2DM rats to investigate whether AS-IV could alleviate diabetic liver injury and to identify its underlying mechanisms. The results demonstrated that AS-IV treatment could restore changes in food intake, water intake, urine volume and body weight, as well as improve liver function and glucose homeostasis in T2DM rats. Moreover, AS-IV treatment promoted suppressed autophagy in the liver of T2DM rats and improved IR, dyslipidaemia, oxidative stress and inflammation. In addition, AS-IV activated adenosine monophosphate-activated protein kinase (AMPK), which inhibited mTOR. Taken together, the present study suggested that AS-IV alleviated diabetic liver injury in T2DM rats, and its mechanism may be associated with the promotion of AMPK/mTOR-mediated autophagy, which further improved IR, dyslipidaemia, oxidative stress and inflammation. Thus, the regulation of autophagy may be an effective strategy to treat diabetic liver injury in T2DM.
Collapse
Affiliation(s)
- Yunfeng Zhu
- Key Laboratory of Anti‑Inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yong Su
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Jie Zhang
- Key Laboratory of Anti‑Inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yanhua Zhang
- Key Laboratory of Anti‑Inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yan Li
- Key Laboratory of Anti‑Inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuli Han
- Key Laboratory of Anti‑Inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xianan Dong
- Key Laboratory of Anti‑Inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Weizu Li
- Key Laboratory of Anti‑Inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Weiping Li
- Key Laboratory of Anti‑Inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
25
|
Reho JJ, Guo DF, Rahmouni K. Mechanistic Target of Rapamycin Complex 1 Signaling Modulates Vascular Endothelial Function Through Reactive Oxygen Species. J Am Heart Assoc 2020; 8:e010662. [PMID: 31020916 PMCID: PMC6512105 DOI: 10.1161/jaha.118.010662] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background The mechanistic target of rapamycin complex 1 (mTORC1) is an important intracellular energy sensor that regulates gene expression and protein synthesis through its downstream signaling components, the S6‐kinase and the ribosomal S6 protein. Recently, signaling arising from mTORC1 has been implicated in regulation of the cardiovascular system with implications for disease. Here, we examined the contribution of mTORC1 signaling to the regulation of vascular function. Methods and Results Activation of mTORC1 pathway in aortic rings with leucine or an adenoviral vector expressing a constitutively active S6‐kinase reduces endothelial‐dependent vasorelaxation in an mTORC1‐dependent manner without affecting smooth muscle relaxation responses. Moreover, activation of mTORC1 signaling in endothelial cells increases reactive oxygen species (ROS) generation and ROS gene expression resulting in a pro‐oxidant gene environment. Blockade of ROS signaling with Tempol restores endothelial function in vascular rings with increased mTORC1 activity indicating a crucial interaction between mTORC1 and ROS signaling. We then tested the role of nuclear factor‐κB transcriptional complex in connecting mTORC1 and ROS signaling in endothelial cells. Blockade of inhibitor of nuclear factor κ‐B kinase subunit β activity with BMS‐345541 prevented the increased ROS generation associated with increased mTORC1 activity in endothelial cells but did not improve vascular endothelial function in aortic rings with increased mTORC1 and ROS signaling. Conclusions These results implicate mTORC1 as a critical molecular signaling hub in the vascular endothelium in mediating vascular endothelial function through modulation of ROS signaling.
Collapse
Affiliation(s)
- John J Reho
- 1 Department of Pharmacology University of Iowa Carver College of Medicine Iowa City IA
| | - Deng-Fu Guo
- 1 Department of Pharmacology University of Iowa Carver College of Medicine Iowa City IA
| | - Kamal Rahmouni
- 1 Department of Pharmacology University of Iowa Carver College of Medicine Iowa City IA.,2 Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,3 Fraternal Order of Eagles Diabetes Research Center University of Iowa Iowa City IA
| |
Collapse
|
26
|
Zhu L, Hu B, Guo Y, Yang H, Zheng J, Yao X, Hu H, Liu H. Effect of Chitosan oligosaccharides on ischemic symptom and gut microbiota disbalance in mice with hindlimb ischemia. Carbohydr Polym 2020; 240:116271. [PMID: 32475560 DOI: 10.1016/j.carbpol.2020.116271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/16/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
This study was designed to explore the effect of Chitosan oligosaccharides (COS) on mouse hindlimb ischemia by femoral artery ligation. Here, we demonstrated that COS treatment statistically promoted the blood perfusion and neovascularization in ischemic hindlimb of mice, accompanied by the suppression of inflammation and oxidative stress. By 16S rDNA gene sequencing, the disbalanced gut microbiota was observed in ischemic mice, while COS treatment, at least in part, restored the abundance changes of some intestinal bacteria at either phylum or genus levels. Based on metabolomics analysis on mouse plasma by UPLC-QTOF-MS, we screened 20 metabolites with the largest responses to ischemia, several of which were markedly reversed by COS. By Spearman's correlation analysis, the changed metabolites might act as a bridge between improved intestinal bacterial structure and alleviated hindlimb ischemia of mice treated by COS. Our studies point towards a potential role of COS in treatment of peripheral ischemia diseases.
Collapse
Affiliation(s)
- Lin Zhu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Baifei Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Yanlei Guo
- Chongqing Academy of Chinese Materia Medica, Nanshan Road 34, Chongqing 400065, PR China
| | - Huabing Yang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Junping Zheng
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Xiaowei Yao
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Haiming Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China.
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Chongqing Academy of Chinese Materia Medica, Nanshan Road 34, Chongqing 400065, PR China.
| |
Collapse
|
27
|
Fan W, Zhang R, Han D, Jiang Z, Li S, Zhang J, Li Y, Wang Y, Cao F. Reduced Sirtuin1 signalling exacerbates diabetic mice hindlimb ischaemia injury and inhibits the protective effect of a liver X receptor agonist. J Cell Mol Med 2020; 24:5476-5490. [PMID: 32286000 PMCID: PMC7214142 DOI: 10.1111/jcmm.15201] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/29/2020] [Accepted: 03/10/2020] [Indexed: 12/30/2022] Open
Abstract
Diabetes mellitus causes endothelial dysfunction, which further exacerbates peripheral arterial disease (PAD). Improving endothelial function via reducing endothelial oxidative stress (OS) may be a promising therapy for diabetic PAD. Activation of liver X receptor (LXR) inhibits excessive OS and provides protective effects on endothelial cells in diabetic individuals. Therefore, we investigated the effects of LXR agonist treatment on diabetic PAD with a focus on modulating endothelial OS. We used a streptozotocin‐induced diabetes mouse model combined with a hindlimb ischaemia (HLI) injury to mimic diabetic PAD, which was followed by LXR agonist treatment. In our study, the LXR agonist T0901317 protected against HLI injury in diabetic mice by attenuating endothelial OS and stimulating angiogenesis. However, a deficiency in endothelial Sirtuin1 (SIRT1) largely inhibited the therapeutic effects of T0901317. Furthermore, we found that the underlying therapeutic mechanisms of T0901317 were related to SIRT1 and non‐SIRT1 signalling, and the isoform LXRβ was involved in LXR agonist‐elicited SIRT1 regulation. In conclusion, LXR agonist treatment protected against HLI injury in diabetic mice via mitigating endothelial OS and stimulating cellular viability and angiogenesis by LXRβ, which elicited both SIRT1‐mediated and non‐SIRT1‐mediated signalling pathways. Therefore, LXR agonist treatment may be a promising therapeutic strategy for diabetic PAD.
Collapse
Affiliation(s)
- Wensi Fan
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases2nd Medical CenterChinese PLA General HospitalBeijingChina
| | - Ran Zhang
- Department of Cardiology1st Medical CenterChinese PLA General HospitalBeijingChina
| | - Dong Han
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXianChina
| | - Zhenhua Jiang
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXianChina
| | - Shuang Li
- Department of CardiologyThe General Hospital of Western Theater Command (Chengdu Military General Hospital)Chengdu610083China
| | - Jibin Zhang
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases2nd Medical CenterChinese PLA General HospitalBeijingChina
| | - Yanhua Li
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases2nd Medical CenterChinese PLA General HospitalBeijingChina
| | - Yabin Wang
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases2nd Medical CenterChinese PLA General HospitalBeijingChina
- Department of Cardiology1st Medical CenterChinese PLA General HospitalBeijingChina
| | - Feng Cao
- Department of Cardiology & National Clinical Research Center for Geriatric Diseases2nd Medical CenterChinese PLA General HospitalBeijingChina
- Department of Cardiology1st Medical CenterChinese PLA General HospitalBeijingChina
| |
Collapse
|
28
|
Kocak Z, Temiz-Resitoglu M, Guden DS, Vezir O, Sucu N, Balcı S, Tamer-Gumus L, Tunctan B, Malik KU, Sahan-Firat S. Modulation of oxidative–nitrosative stress and inflammatory response by rapamycin in target and distant organs in rats exposed to hindlimb ischemia–reperfusion: the role of mammalian target of rapamycin. Can J Physiol Pharmacol 2019; 97:1193-1203. [DOI: 10.1139/cjpp-2019-0394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Mammalian target of rapamycin (mTOR) has been recognized with potential immunomodulatory properties playing an important role in various physiopathological processes including ischemia–reperfusion (I/R) injury. I/R injury stimulate reactive oxygen and nitrogen species by activating nicotinamide adenine dinucleotide phosphate oxidase and inducible nitric oxide synthase, respectively. Controversial results have been obtained in different I/R models following localized I/R; however, the precise role of the mTOR signaling pathway remains undefined. The objective of the current study was to evaluate the role of the mTOR in oxidative–nitrosative stress and inflammation in hindlimb I/R-induced injury in target and remote organ injuries. In rats subjected to I/R, an increased expression of ribosomal protein S6 (rpS6), inhibitor κB (IκB)-α, nuclear factor-κB (NF-κB) p65, inducible nitric oxide synthase, cyclooxygenase 2, gp91phox, and levels of tumor necrosis factor α, nitrite, nitrotyrosine, malondialdehyde and the activities of myeloperoxidase and catalase in the tissues and (or) sera were detected. Treatment with rapamycin, a selective inhibitor of mTOR, reversed all the I/R-induced changes as manifested by its anti-inflammatory and antioxidant effects in kidney and gastrocnemius muscle of rats. Collectively, these findings suggest that rapamycin protects against I/R-induced oxidative–nitrosative stress and inflammation leading to organ injuries via suppression of mTOR/IκB-α/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zumrut Kocak
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, 33169 Mersin, Turkey
| | - Meryem Temiz-Resitoglu
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, 33169 Mersin, Turkey
| | - Demet Sinem Guden
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, 33169 Mersin, Turkey
| | - Ozden Vezir
- Department of Cardiovascular Surgery, Mersin State Hospital, 33240 Mersin, Turkey
| | - Nehir Sucu
- Department of Cardiovascular Surgery, Faculty of Medicine, Mersin University, 33150 Mersin, Turkey
| | - Senay Balcı
- Department of Biochemistry, Faculty of Medicine, Mersin University, 33150 Mersin, Turkey
| | - Lulufer Tamer-Gumus
- Department of Biochemistry, Faculty of Medicine, Mersin University, 33150 Mersin, Turkey
| | - Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, 33169 Mersin, Turkey
| | - Kafait U. Malik
- Department of Pharmacology, College of Medicine, University of Tennessee, Center for Health Sciences, Memphis, TN 38163, USA
| | - Seyhan Sahan-Firat
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, 33169 Mersin, Turkey
| |
Collapse
|
29
|
Kobialka P, Graupera M. Revisiting PI3-kinase signalling in angiogenesis. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2019; 1:H125-H134. [PMID: 32923964 PMCID: PMC7439845 DOI: 10.1530/vb-19-0025] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022]
Abstract
PI3Ks belong to a family of lipid kinases that comprises eight isoforms. They phosphorylate the third position of the inositol ring present in phosphatidylinositol lipids and, in turn, activate a broad range of proteins. The PI3K pathway regulates primal cellular responses, including proliferation, migration, metabolism and vesicular traffic. These processes are fundamental for endothelial cell function during sprouting angiogenesis, the most common type of blood vessel formation. Research in animal models has revealed key functions of PI3K family members and downstream effectors in angiogenesis. In addition, perturbations in PI3K signalling have been associated with aberrant vascular growth including tumour angiogenesis and vascular malformations. Together, this highlights that endothelial cells are uniquely sensitive to fluctuations in PI3K signalling. Here, we aim to update the current view on this important signalling cue in physiological and pathological blood vessel growth.
Collapse
Affiliation(s)
- Piotr Kobialka
- Vascular Biology and Signalling Group, Program Against Cancer Therapeutic Resistance (ProCURE), Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat-Barcelona, Spain
- ProCure Research Program, Instituto de Salud Carlos III, Madrid, Spain
- OncoBell Program, Instituto de Salud Carlos III, Madrid, Spain
| | - Mariona Graupera
- Vascular Biology and Signalling Group, Program Against Cancer Therapeutic Resistance (ProCURE), Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat-Barcelona, Spain
- ProCure Research Program, Instituto de Salud Carlos III, Madrid, Spain
- OncoBell Program, Instituto de Salud Carlos III, Madrid, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
30
|
CTRP3 Protects against High Glucose-Induced Cell Injury in Human Umbilical Vein Endothelial Cells. Anal Cell Pathol (Amst) 2019; 2019:7405602. [PMID: 31428552 PMCID: PMC6681575 DOI: 10.1155/2019/7405602] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/12/2019] [Indexed: 12/23/2022] Open
Abstract
Aims Inflammation was closely associated with diabetes-related endothelial dysfunction. C1q/tumor necrosis factor-related protein 3 (CTRP3) is a member of the CTRP family and can provide cardioprotection in many cardiovascular diseases via suppressing the production of inflammatory factors. However, the role of CTRP3 in high glucose- (HG-) related endothelial dysfunction remains unclear. This study evaluates the effects of CTRP3 on HG-induced cell inflammation and apoptosis. Materials and Methods To prevent high glucose-induced cell injury, human umbilical vein endothelial cells (HUVECs) were pretreated with recombinant CTRP3 for 1 hour followed by normal glucose (5.5 mmol/l) or high glucose (33 mmol/l) treatment. After that, cell apoptosis and inflammatory factors were determined. Results Our results demonstrated that CTRP3 mRNA and protein expression were significantly decreased after HG exposure in HUVECs. Recombinant human CTRP3 inhibited HG-induced accumulation of inflammatory factors and cell loss in HUVECs. CTRP3 treatment also increased the phosphorylation levels of protein kinase B (AKT/PKB) and the mammalian target of rapamycin (mTOR) in HUVECs. CTRP3 lost its inhibitory effects on HG-induced cell inflammation and apoptosis after AKT inhibition. Knockdown of endogenous CTRP3 in HUVECs resulted in increased inflammation and decreased cell viability in vitro. Conclusions Taken together, these findings indicated that CTRP3 treatment blocked the accumulation of inflammatory factors and cell loss in HUVECs after HG exposure through the activation of AKT-mTOR signaling pathway. Thus, CTRP3 may be a potential therapeutic drug for the prevention of diabetes-related endothelial dysfunction.
Collapse
|
31
|
Liu D, Xu L, Zhang X, Shi C, Qiao S, Ma Z, Yuan J. Snapshot: Implications for mTOR in Aging-related Ischemia/Reperfusion Injury. Aging Dis 2019; 10:116-133. [PMID: 30705773 PMCID: PMC6345330 DOI: 10.14336/ad.2018.0501] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 05/01/2018] [Indexed: 12/15/2022] Open
Abstract
Aging may aggravate the damage and dysfunction of different components of multiorgan and thus increasing multiorgan ischemia/reperfusion (IR) injury. IR injury occurs in many organs and tissues, which is a major cause of morbidity and mortality worldwide. The kinase mammalian target of rapamycin (mTOR), an atypical serine/threonine protein kinase, involves in the pathophysiological process of IR injury. In this review, we first briefly introduce the molecular features of mTOR, the association between mTOR and aging, and especially its role on autophagy. Special focus is placed on the roles of mTOR during ischemic and IR injury. We then clarify the association between mTOR and conditioning phenomena. Following this background, we expand our discussion to potential future directions of research in this area. Collectively, information reviewed herein will serve as a comprehensive reference for the actions of mTOR in IR injury and may be significant for the design of future research and increase the potential of mTOR as a therapeutic target.
Collapse
Affiliation(s)
- Dong Liu
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Liqun Xu
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.,2Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China.,3Cadet group 3, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032, China.,4Laboratory Animal Center, The Fourth Military Medical University, Xi'an 710032, China
| | - Xiaoyan Zhang
- 2Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China.,3Cadet group 3, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032, China
| | - Changhong Shi
- 4Laboratory Animal Center, The Fourth Military Medical University, Xi'an 710032, China
| | - Shubin Qiao
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zhiqiang Ma
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.,2Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Jiansong Yuan
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| |
Collapse
|
32
|
Sun Z, Ma N, Fan W, Guo L, Chen J, Zhu L, Tong G. Noninvasive monitoring of the development and treatment response of ischemic hindlimb by targeting matrix metalloproteinase-2 (MMP-2). Biomater Sci 2019; 7:4036-4045. [PMID: 31482934 DOI: 10.1039/c9bm00915a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
An ultrafast MMP activatable probe monitoring the development of angiogenesis in ischemic hindlimb through fluorescence imaging in real-time.
Collapse
Affiliation(s)
- Zhongchan Sun
- Department of Cardiology
- Guangdong Cardiovascular Institute
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease
- Guangdong Provincial People's Hospital
- Guangdong Academy of Medical Sciences
| | - Nan Ma
- Department of Ophthalmology
- Tangdu Hospital
- Fourth Military Medical University
- Xi'an
- China
| | - Wensi Fan
- Department of Cardiology
- Xijing Hospital
- Fourth Military Medical University
- Xi'an
- China
| | - Lanyan Guo
- Department of Cardiology
- Xijing Hospital
- Fourth Military Medical University
- Xi'an
- China
| | - Jiangwei Chen
- Department of Cardiology
- Xijing Hospital
- Fourth Military Medical University
- Xi'an
- China
| | - Lei Zhu
- Winship Cancer Institute
- Department of Surgery
- Emory University
- Atlanta
- USA
| | - Guang Tong
- Department of Cardiac Surgery
- Guangdong Cardiovascular Institute
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease
- Guangdong Provincial People's Hospital
- Guangdong Academy of Medical Sciences
| |
Collapse
|
33
|
Sun C, Diao Q, Lu J, Zhang Z, Wu D, Wang X, Xie J, Zheng G, Shan Q, Fan S, Hu B, Zheng Y. Purple sweet potato color attenuated NLRP3 inflammasome by inducing autophagy to delay endothelial senescence. J Cell Physiol 2018; 234:5926-5939. [DOI: 10.1002/jcp.28003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 06/13/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Chunhui Sun
- Institute for Advanced Interdisciplinary Research, University of Jinan Jinan China
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Qiaoqiao Diao
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Zifeng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Dongmei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Xingqi Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Jun Xie
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Guihong Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Shaohua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Yuanlin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| |
Collapse
|
34
|
Chen L, Zhang L, Fang Z, Li C, Yang Y, You X, Song M, Coffie J, Zhang L, Gao X, Wang H. Naoxintong restores collateral blood flow in a murine model of hindlimb ischemia through PPARδ-dependent mechanism. JOURNAL OF ETHNOPHARMACOLOGY 2018; 227:121-130. [PMID: 30170078 DOI: 10.1016/j.jep.2018.08.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 07/04/2018] [Accepted: 08/26/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Naoxintong (NXT) is a compound preparation that is widely used in patients with cardiovascular and cerebrovascular diseases. AIM OF STUDY The aim of this study is to investigate the protective mechanism of NXT on the mice model of peripheral vascular disease (PAD). MATERIALS AND METHODS In the study, hindlimb ischemia was induced by ligation of femoral artery on the right leg of mice. After surgery, the mice were administrated with saline solution, 10 mg/kg/d simvastatin and 700 mg/kg/d NXT for 4 weeks. The blood flow perfusion was measured by laser Doppler perfusion imaging system. Histological and immunofluorescent staining was used to determine muscle recovery, capillary density, tissue vascular endothelial growth factor (VEGF), phosphorylated-Akt (p-Akt) and phosphorylated-endothelial nitric oxide synthase (p-eNOS) expression. Terminal deoxynucleotidyl transferased UTP nick end labeling (TUNEL) was performed to detect the apoptosis of myocytes in hindlimb. The autophagy-associated gene expression and peroxisome proliferator-activated receptors (PPARs) expression were measured by Quantitative Real-Time Reverse Transcription Polymerase Chain Reaction (qRT-PCR). Western blotting was performed to detect the expressions of light-chain 3 (LC3), VEGF, p-Akt, p-eNOS and PPARs. The EMSA experiment was performed to figure out whether PPARδ could directly bind to the predicted PPRE motif of VEGF. RESULTS NXT treatment significantly accelerated perfusion recovery and reduced tissue injury in mice muscle. Apoptosis and autophagy were decreased within the ischemic muscle of NXT-treated mice. Quantification of vessels in hindlimb muscles provided evidences that NXT promoted angiogenesis in peripheral ischemia. In addition, results from western blotting and immunofluorescent staining suggested NXT induced angiogenesis via VEGF/Akt/eNOS signaling pathway. More interestingly, NXT specifically increased the expression of PPARδ in both mRNA and protein levels. EMSA results showed that PPARδ associated with PPRE site of VEGF promoter, suggesting that NXT-induced VEGF expression is mediated, at least in part, by PPARδ. CONCLUSION In conclusion, the present study implicated that the restoration of hindlimb blood perfusion and recovery of limb functions were improved in NXT-treated mice with significant improvement of angiogenesis mediated by PPARδ-VEGF-Akt-eNOS axis as well as attenuation of autophagy and apoptosis. These results expand knowledge about the beneficial effects of NXT in angiogenesis and blood flow recovery. It might provide insight into the PPARδ regulating neovascularization in hindlimb ischemia and identify NXT as a potent new compound used for the treatment of peripheral vascular disease.
Collapse
Affiliation(s)
- Lu Chen
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Lusha Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Zhirui Fang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Chunxiao Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yue Yang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin 300193, China.
| | - Xingyu You
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Min Song
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Joel Coffie
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Liyuan Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Hong Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
35
|
Jiang C, Jiang L, Li Q, Liu X, Zhang T, Dong L, Liu T, Liu L, Hu G, Sun X, Jiang L. Acrolein induces NLRP3 inflammasome-mediated pyroptosis and suppresses migration via ROS-dependent autophagy in vascular endothelial cells. Toxicology 2018; 410:26-40. [DOI: 10.1016/j.tox.2018.09.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/31/2018] [Accepted: 09/06/2018] [Indexed: 12/20/2022]
|
36
|
Liu R, Luo Q, You W, Jin M. MicroRNA-106 attenuates hyperglycemia-induced vascular endothelial cell dysfunction by targeting HMGB1. Gene 2018; 677:142-148. [DOI: 10.1016/j.gene.2018.07.063] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/22/2018] [Accepted: 07/24/2018] [Indexed: 01/13/2023]
|
37
|
Zhang Y, Ma KL, Gong YX, Wang GH, Hu ZB, Liu L, Lu J, Chen PP, Lu CC, Ruan XZ, Liu BC. Platelet Microparticles Mediate Glomerular Endothelial Injury in Early Diabetic Nephropathy. J Am Soc Nephrol 2018; 29:2671-2695. [PMID: 30341150 DOI: 10.1681/asn.2018040368] [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: 04/09/2018] [Accepted: 09/18/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Glomerular endothelium dysfunction, which plays a crucial role in the pathogenesis of early diabetic nephropathy, might be caused by circulating metabolic abnormalities. Platelet microparticles, extracellular vesicles released from activated platelets, have recently emerged as a novel regulator of vascular dysfunction. METHODS We studied the effects of platelet microparticles on glomerular endothelial injury in early diabetic nephropathy in rats with streptozotocin-induced diabetes and primary rat glomerular endothelial cells. Isolated platelet microparticles were measured by flow cytometry. RESULTS Plasma platelet microparticles were significantly increased in diabetic rats, an effect inhibited in aspirin-treated animals. In cultured glomerular endothelial cells, platelet microparticles induced production of reactive oxygen species, decreased nitric oxide levels, inhibited activities of endothelial nitric oxide synthase and SOD, increased permeability of the glomerular endothelium barrier, and reduced thickness of the endothelial surface layer. Conversely, inhibition of platelet microparticles in vivo by aspirin improved glomerular endothelial injury. Further analysis showed that platelet microparticles activated the mammalian target of rapamycin complex 1 (mTORC1) pathway in glomerular endothelial cells; inhibition of the mTORC1 pathway by rapamycin or raptor siRNA significantly protected against microparticle-induced glomerular endothelial injury in vivo and in vitro. Moreover, platelet microparticle-derived chemokine ligand 7 (CXCL7) contributed to glomerular endothelial injury, and antagonizing CXCL7 using CXCL7-neutralizing antibody or blocking CXCL7 receptors with a competitive inhibitor of CXCR1 and CXCR2 dramatically attenuated such injury. CONCLUSIONS These findings demonstrate a pathogenic role of platelet microparticles in glomerular endothelium dysfunction, and suggest a potential therapeutic target, CXCL7, for treatment of early diabetic nephropathy.
Collapse
Affiliation(s)
- Yang Zhang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; and
| | - Kun Ling Ma
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; and
| | - Yu Xiang Gong
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; and
| | - Gui Hua Wang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; and
| | - Ze Bo Hu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; and
| | - Liang Liu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; and
| | - Jian Lu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; and
| | - Pei Pei Chen
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; and
| | - Chen Chen Lu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; and
| | - Xiong Zhong Ruan
- Centre for Nephrology, University College London Medical School, London, UK
| | - Bi Cheng Liu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; and
| |
Collapse
|
38
|
Autophagy and Akt in the protective effect of erythropoietin helix B surface peptide against hepatic ischaemia/reperfusion injury in mice. Sci Rep 2018; 8:14703. [PMID: 30279567 PMCID: PMC6168561 DOI: 10.1038/s41598-018-33028-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 09/17/2018] [Indexed: 12/27/2022] Open
Abstract
Helix B surface peptide (HBSP) is an erythropoietin (EPO)-derived peptide that protects tissue from the risks of elevated blood pressure and thrombosis. This study focused on the protection of HBSP in hepatic ischaemia/reperfusion (I/R) by enhancing the level of autophagy. In detail, we randomly divided C57BL/6 mice into sham-operated, hepatic ischaemia/reperfusion (I/R), I/R + HBSP, I/R + HBSP + 3-methyladenine (autophagy inhibitor), I/R + HBSP + rapamycin (mTOR inhibitor), and I/R + HBSP + Ly294002 (Akt inhibitor) groups. We assessed alanine aminotransferase (ALT), aspartate aminotransferase (AST) and lactate dehydrogenase (LDH) levels in mouse sera, and performed haematoxylin/eosin (HE) staining, immunohistochemistry, electron microscopy, immunofluorescence microscopy, and western blotting on liver tissue to detect the degree of liver injury, liver apoptosis, autophagy, and the expression of microtubule associated protein 1 light chain 3 alpha (Map1lc3, or LC3), Beclin 1, phospho-mTOR, mTOR, phospho-Akt (P-Akt), and Akt. HBSP relieved hepatic I/R injury in a concentration-independent manner. The expression of LC3II, LC3I, and Beclin 1, and the formation of autophagosomes, in the I/R + HBSP group were higher than those in the I/R group. The protective effects of HBSP were abolished by 3-methyladenine and, to a lesser extent, Ly294002, but enhanced by rapamycin. Furthermore, In vivo, HBSP also protected against hypoxia injury induced by cobalt chloride (CoCl2) through improving the level of autophagy. Therefore, HBSP protected against hepatic I/R injury, mainly via regulating autophagy by targeting mTOR.
Collapse
|
39
|
Haissaguerre M, Ferrière A, Simon V, Saucisse N, Dupuy N, André C, Clark S, Guzman-Quevedo O, Tabarin A, Cota D. mTORC1-dependent increase in oxidative metabolism in POMC neurons regulates food intake and action of leptin. Mol Metab 2018; 12:98-106. [PMID: 29699927 PMCID: PMC6001919 DOI: 10.1016/j.molmet.2018.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/05/2018] [Accepted: 04/07/2018] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVE Nutrient availability modulates reactive oxygen species (ROS) production in the hypothalamus. In turn, ROS regulate hypothalamic neuronal activity and feeding behavior. The mechanistic target of rapamycin complex 1 (mTORC1) pathway is an important cellular integrator of the action of nutrients and hormones. Here we tested the hypothesis that modulation of mTORC1 activity, particularly in Proopiomelanocortin (POMC)-expressing neurons, mediates the cellular and behavioral effects of ROS. METHODS C57BL/6J mice or controls and their knockout (KO) littermates deficient either for the mTORC1 downstream target 70-kDa ribosomal protein S6 kinase 1 (S6K1) or for the mTORC1 component Rptor specifically in POMC neurons (POMC-rptor-KO) were treated with an intracerebroventricular (icv) injection of the ROS hydrogen peroxide (H2O2) or the ROS scavenger honokiol, alone or, respectively, in combination with the mTORC1 inhibitor rapamycin or the mTORC1 activator leptin. Oxidant-related signal in POMC neurons was assessed using dihydroethidium (DHE) fluorescence. RESULTS Icv administration of H2O2 decreased food intake, while co-administration of rapamycin, whole-body deletion of S6K1, or deletion of rptor in POMC neurons impeded the anorectic action of H2O2. H2O2 also increased oxidant levels in POMC neurons, an effect that hinged on functional mTORC1 in these neurons. Finally, scavenging ROS prevented the hypophagic action of leptin, which in turn required mTORC1 to increase oxidant levels in POMC neurons and to inhibit food intake. CONCLUSIONS Our results demonstrate that ROS and leptin require mTORC1 pathway activity in POMC neurons to increase oxidant levels in POMC neurons and consequently decrease food intake.
Collapse
Affiliation(s)
- Magalie Haissaguerre
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; Department of Endocrinology, Hôpital Haut Lévèque, CHU Bordeaux, F-33600 Pessac, France
| | - Amandine Ferrière
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; Department of Endocrinology, Hôpital Haut Lévèque, CHU Bordeaux, F-33600 Pessac, France
| | - Vincent Simon
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France
| | - Nicolas Saucisse
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France
| | - Nathalie Dupuy
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France
| | - Caroline André
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France
| | - Samantha Clark
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France
| | - Omar Guzman-Quevedo
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France
| | - Antoine Tabarin
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; Department of Endocrinology, Hôpital Haut Lévèque, CHU Bordeaux, F-33600 Pessac, France
| | - Daniela Cota
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France.
| |
Collapse
|
40
|
Fan Y, Lu H, Liang W, Garcia-Barrio MT, Guo Y, Zhang J, Zhu T, Hao Y, Zhang J, Chen YE. Endothelial TFEB (Transcription Factor EB) Positively Regulates Postischemic Angiogenesis. Circ Res 2018; 122:945-957. [PMID: 29467198 DOI: 10.1161/circresaha.118.312672] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/11/2018] [Accepted: 02/19/2018] [Indexed: 12/25/2022]
Abstract
RATIONALE Postischemic angiogenesis is critical to limit the ischemic tissue damage and improve the blood flow recovery. The regulation and the underlying molecular mechanisms of postischemic angiogenesis are not fully unraveled. TFEB (transcription factor EB) is emerging as a master gene for autophagy and lysosome biogenesis. However, the role of TFEB in vascular disease is less understood. OBJECTIVE We aimed to determine the role of endothelial TFEB in postischemic angiogenesis and its underlying molecular mechanism. METHODS AND RESULTS In primary human endothelial cells (ECs), serum starvation induced TFEB nuclear translocation. VEGF (vascular endothelial growth factor) increased TFEB expression level and nuclear translocation. Utilizing genetically engineered EC-specific TFEB transgenic and KO (knockout) mice, we investigated the role of TFEB in postischemic angiogenesis in the mouse hindlimb ischemia model. We observed improved blood perfusion and increased capillary density in the EC-specific TFEB transgenic mice compared with the wild-type littermates. Furthermore, blood flow recovery was attenuated in EC-TFEB KO mice compared with control mice. In aortic ring cultures, the TFEB transgene significantly increased vessel sprouting, whereas TFEB deficiency impaired the vessel sprouting. In vitro, adenovirus-mediated TFEB overexpression promoted EC tube formation, migration, and survival, whereas siRNA-mediated TFEB knockdown had the opposite effect. Mechanistically, TFEB activated AMPK (AMP-activated protein kinase)-α signaling and upregulated autophagy. Through inactivation of AMPKα or inhibition of autophagy, we demonstrated that the AMPKα and autophagy are necessary for TFEB to regulate angiogenesis in ECs. Finally, the positive effect of TFEB on AMPKα activation and EC tube formation was mediated by TFEB-dependent transcriptional upregulation of MCOLN1 (mucolipin-1). CONCLUSIONS In summary, our data demonstrate that TFEB is a positive regulator of angiogenesis through activation of AMPKα and autophagy, suggesting that TFEB constitutes a novel molecular target for ischemic vascular disease.
Collapse
Affiliation(s)
- Yanbo Fan
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor.
| | - Haocheng Lu
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor
| | - Wenying Liang
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor
| | - Minerva T Garcia-Barrio
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor
| | - Yanhong Guo
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor
| | - Ji Zhang
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor
| | - Tianqing Zhu
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor
| | - Yibai Hao
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor
| | - Jifeng Zhang
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor
| | - Y Eugene Chen
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor.
| |
Collapse
|
41
|
Tian H, Li S, Yu K. DJ‑1 alleviates high glucose‑induced endothelial cells injury via PI3K/Akt‑eNOS signaling pathway. Mol Med Rep 2017; 17:1205-1211. [PMID: 29115508 DOI: 10.3892/mmr.2017.7975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 10/13/2017] [Indexed: 11/06/2022] Open
Abstract
Hyperglycemia mediated endothelial cells (ECs) injury is closely associated with diabetic vascular complications. It was revealed that DJ‑1 possesses cellular protective effects by suppressing oxidative stress. The present study aimed to investigate the beneficial effects of DJ‑1 on high glucose (HG)‑induced human umbilical vein endothelial cell (HUVEC) injury and to elucidate its underlying mechanisms. HUVECs were incubated under 5.5 mM (control group) or 25 mM D‑glucose (HG group) and then transfected with recombinant adenoviral vectors to overexpression of DJ‑1. Cell proliferation and apoptosis were measured using the EdU incorporation assay and flow cytometry with Annexin V-FITC/propidium iodide double staining, respectively. Apoptotic‑related proteins were determined using western blot analysis. Reactive oxygen species (ROS) production, lactate dehydrogenase (LDH) and nitric oxide (NO) levels, the content of malondialdehyde (MDA), and the activities of superoxide dismutase (SOD) were measured. Results demonstrated that overexpression of DJ‑1 promoted cell proliferation and inhibited HUVECs apoptosis stimulated by HG. DJ‑1 also suppressed the HG‑induced reduction in the Bcl‑2/Bax ratio and HG activated ROS generation in HUVECs. Furthermore, HG significantly increased the levels of LDH and MDA, and reduced the level of SOD; however, these effects were reversed by Ad‑DJ‑1 transfection. Furthermore, the cellular protective effect of overexpression of DJ‑1 enhanced p‑Akt/Akt ratio, eNOS activation and NO production, and these trends were partially reversed by a phosphatidylinositol‑4,5‑bisphosphate 3‑kinase (PI3K) inhibitor (LY294002). Taken together, the present study highlighted the involvement of DJ‑1 in HG‑related EC injury and identified that DJ‑1 exerts a cellular protective effect in HUVECs exposed to HG induced oxidative stress via activation of the PI3K/Akt‑eNOS signaling pathway.
Collapse
Affiliation(s)
- Hongan Tian
- Radiology Department, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Shunzhen Li
- Radiology Department, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Kaihu Yu
- Radiology Department, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| |
Collapse
|