1
|
Guo L, Zong Y, Yang W, Lin Y, Feng Q, Yu C, Liu X, Li C, Zhang W, Wang R, Li L, Pei Y, Wang H, Liu D, Niu H, Nie L. DCBLD2 deletion increases hyperglycemia and induces vascular remodeling by inhibiting insulin receptor recycling in endothelial cells. FEBS J 2024; 291:4076-4095. [PMID: 38872483 DOI: 10.1111/febs.17198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 04/02/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
Discoidin, CUB, LCCL domain-containing 2 (DCBLD2) is a type I transmembrane protein with a similar structure to neuropilin, which acts as a co-receptor for certain receptor tyrosine kinases (RTKs). The insulin receptor is an RTK and plays a critical role in endothelial cell function and glycolysis. However, how and whether DCBLD2 regulates insulin receptor activity in endothelial cells is poorly understood. Diabetes was induced through treatment of Dcbld2 global-genome knockout mice and endothelium-specific knockout mice with streptozotocin. Vascular ultrasound, vascular tension test, and hematoxylin and eosin staining were performed to assess endothelial function and aortic remodeling. Glycolytic rate assays, real-time PCR and western blotting were used to investigate the effects of DCBLD2 on glycolytic activity and insulin receptor (InsR)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway in endothelial cells. Co-immunoprecipitation was used to assess the effects of DCBLD2 on insulin receptor endocytosis and recycling. Membrane and cytoplasmic proteins were isolated to determine whether DCBLD2 could affect the localization of the insulin receptor. We found that Dcbld2 deletion exacerbated endothelial dysfunction and vascular remodeling in diabetic mice. Both Dcbld2 knockdown and Dcbld2 deletion inhibited glycolysis and the InsR/PI3K/Akt signaling pathway in endothelial cells. Furthermore, Dcbld2 deletion inhibited insulin receptor recycling. Taken together, Dcbld2 deficiency exacerbated diabetic endothelial dysfunction and vascular remodeling by inhibiting the InsR/PI3K/Akt pathway in endothelial cells through the inhibition of Rab11-dependent insulin receptor recycling. Our data suggest that DCBLD2 is a potential therapeutic target for diabetes and cardiovascular diseases.
Collapse
Affiliation(s)
- Lingling Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Yanhong Zong
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Weiwei Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Yanling Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Qi Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Chao Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Xiaoning Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Chenyang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Wenjun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Runtao Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Lijing Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Yunli Pei
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Huifang Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Demin Liu
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Honglin Niu
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
- School of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Lei Nie
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
2
|
Jin H, Zhang C, Nagenborg J, Juhasz P, Ruder AV, Sikkink CJJM, Mees BME, Waring O, Sluimer JC, Neumann D, Goossens P, Donners MMPC, Mardinoglu A, Biessen EAL. Genome-scale metabolic network of human carotid plaque reveals the pivotal role of glutamine/glutamate metabolism in macrophage modulating plaque inflammation and vulnerability. Cardiovasc Diabetol 2024; 23:240. [PMID: 38978031 PMCID: PMC11232311 DOI: 10.1186/s12933-024-02339-3] [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: 10/30/2023] [Accepted: 06/26/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Metabolism is increasingly recognized as a key regulator of the function and phenotype of the primary cellular constituents of the atherosclerotic vascular wall, including endothelial cells, smooth muscle cells, and inflammatory cells. However, a comprehensive analysis of metabolic changes associated with the transition of plaque from a stable to a hemorrhaged phenotype is lacking. METHODS In this study, we integrated two large mRNA expression and protein abundance datasets (BIKE, n = 126; MaasHPS, n = 43) from human atherosclerotic carotid artery plaque to reconstruct a genome-scale metabolic network (GEM). Next, the GEM findings were linked to metabolomics data from MaasHPS, providing a comprehensive overview of metabolic changes in human plaque. RESULTS Our study identified significant changes in lipid, cholesterol, and inositol metabolism, along with altered lysosomal lytic activity and increased inflammatory activity, in unstable plaques with intraplaque hemorrhage (IPH+) compared to non-hemorrhaged (IPH-) plaques. Moreover, topological analysis of this network model revealed that the conversion of glutamine to glutamate and their flux between the cytoplasm and mitochondria were notably compromised in hemorrhaged plaques, with a significant reduction in overall glutamate levels in IPH+ plaques. Additionally, reduced glutamate availability was associated with an increased presence of macrophages and a pro-inflammatory phenotype in IPH+ plaques, suggesting an inflammation-prone microenvironment. CONCLUSIONS This study is the first to establish a robust and comprehensive GEM for atherosclerotic plaque, providing a valuable resource for understanding plaque metabolism. The utility of this GEM was illustrated by its ability to reliably predict dysregulation in the cholesterol hydroxylation, inositol metabolism, and the glutamine/glutamate pathway in rupture-prone hemorrhaged plaques, a finding that may pave the way to new diagnostic or therapeutic measures.
Collapse
Affiliation(s)
- Han Jin
- Central Laboratory, Tianjin Medical University General Hospital, Tianjin, China
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht, the Netherlands
- Science for Life Laboratory (SciLifeLab), KTH-Royal Institute of Technology, Solna, Sweden
| | - Cheng Zhang
- Science for Life Laboratory (SciLifeLab), KTH-Royal Institute of Technology, Solna, Sweden
| | - Jan Nagenborg
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht, the Netherlands
| | | | - Adele V Ruder
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht, the Netherlands
| | | | - Barend M E Mees
- Department of Surgery, Maastricht UMC+, Maastricht, the Netherlands
| | - Olivia Waring
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht, the Netherlands
| | - Judith C Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht, the Netherlands
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland
| | - Dietbert Neumann
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht, the Netherlands
| | - Pieter Goossens
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht, the Netherlands
| | - Marjo M P C Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht, the Netherlands
| | - Adil Mardinoglu
- Science for Life Laboratory (SciLifeLab), KTH-Royal Institute of Technology, Solna, Sweden.
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK.
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht, the Netherlands.
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
3
|
Chen X, Xu Y, Ju Y, Gu P. Metabolic Regulation of Endothelial Cells: A New Era for Treating Wet Age-Related Macular Degeneration. Int J Mol Sci 2024; 25:5926. [PMID: 38892113 PMCID: PMC11172501 DOI: 10.3390/ijms25115926] [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: 04/16/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Wet age-related macular degeneration (wet AMD) is a primary contributor to visual impairment and severe vision loss globally, but the prevailing treatments are often unsatisfactory. The development of conventional treatment strategies has largely been based on the understanding that the angiogenic switch of endothelial cells (ECs) is mainly dictated by angiogenic growth factors. Even though treatments targeting vascular endothelial growth factor (VEGF), like ranibizumab, are widely administered, more than half of patients still exhibit inadequate or null responses, suggesting the involvement of other pathogenic mechanisms. With advances in research in recent years, it has become well recognized that EC metabolic regulation plays an active rather than merely passive responsive role in angiogenesis. Disturbances of these metabolic pathways may lead to excessive neovascularization in angiogenic diseases such as wet AMD, therefore targeted modulation of EC metabolism represents a promising therapeutic strategy for wet AMD. In this review, we comprehensively discuss the potential applications of EC metabolic regulation in wet AMD treatment from multiple perspectives, including the involvement of ECs in wet AMD pathogenesis, the major endothelial metabolic pathways, and novel therapeutic approaches targeting metabolism for wet AMD.
Collapse
Affiliation(s)
- Xirui Chen
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (X.C.)
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Yang Xu
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (X.C.)
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Yahan Ju
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (X.C.)
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Ping Gu
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (X.C.)
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| |
Collapse
|
4
|
Liu Y, Wu Z, Li Y, Chen Y, Zhao X, Wu M, Xia Y. Metabolic reprogramming and interventions in angiogenesis. J Adv Res 2024:S2090-1232(24)00178-4. [PMID: 38704087 DOI: 10.1016/j.jare.2024.05.001] [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: 03/15/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Endothelial cell (EC) metabolism plays a crucial role in the process of angiogenesis. Intrinsic metabolic events such as glycolysis, fatty acid oxidation, and glutamine metabolism, support secure vascular migration and proliferation, energy and biomass production, as well as redox homeostasis maintenance during vessel formation. Nevertheless, perturbation of EC metabolism instigates vascular dysregulation-associated diseases, especially cancer. AIM OF REVIEW In this review, we aim to discuss the metabolic regulation of angiogenesis by EC metabolites and metabolic enzymes, as well as prospect the possible therapeutic opportunities and strategies targeting EC metabolism. KEY SCIENTIFIC CONCEPTS OF REVIEW In this work, we discuss various aspects of EC metabolism considering normal and diseased vasculature. Of relevance, we highlight that the implications of EC metabolism-targeted intervention (chiefly by metabolic enzymes or metabolites) could be harnessed in orchestrating a spectrum of pathological angiogenesis-associated diseases.
Collapse
Affiliation(s)
- Yun Liu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zifang Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yikun Li
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yating Chen
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Xuan Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Miaomiao Wu
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Yaoyao Xia
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
5
|
Cao X, Wu VWY, Han Y, Hong H, Wu Y, Kong APS, Lui KO, Tian XY. Role of Argininosuccinate Synthase 1 -Dependent L-Arginine Biosynthesis in the Protective Effect of Endothelial Sirtuin 3 Against Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307256. [PMID: 38233193 DOI: 10.1002/advs.202307256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/08/2023] [Indexed: 01/19/2024]
Abstract
Atherosclerosis is initiated with endothelial cell (EC) dysfunction and vascular inflammation under hyperlipidemia. Sirtuin 3 (SIRT3) is a mitochondrial deacetylase. However, the specific role of endothelial SIRT3 during atherosclerosis remains poorly understood. The present study aims to study the role and mechanism of SIRT3 in EC function during atherosclerosis. Wild-type Sirt3f/f mice and endothelium-selective SIRT3 knockout Sirt3f/f; Cdh5Cre/+ (Sirt3EC-KO) mice are injected with adeno-associated virus (AAV) to overexpress PCSK9 and fed with high-cholesterol diet (HCD) for 12 weeks to induce atherosclerosis. Sirt3EC-KO mice exhibit increased atherosclerotic plaque formation, along with elevated macrophage infiltration, vascular inflammation, and reduced circulating L-arginine levels. In human ECs, SIRT3 inhibition resulted in heightened vascular inflammation, reduced nitric oxide (NO) production, increased reactive oxygen species (ROS), and diminished L-arginine levels. Silencing of SIRT3 results in hyperacetylation and deactivation of Argininosuccinate Synthase 1 (ASS1), a rate-limiting enzyme involved in L-arginine biosynthesis, and this effect is abolished in mutant ASS1. Furthermore, L-arginine supplementation attenuates enhanced plaque formation and vascular inflammation in Sirt3EC-KO mice. This study provides compelling evidence supporting the protective role of endothelial SIRT3 in atherosclerosis and also suggests a critical role of SIRT3-induced deacetylation of ASS1 by ECs for arginine synthesis.
Collapse
Affiliation(s)
- Xiaoyun Cao
- School of Biomedical Sciences, Heart and Vascular Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR, 999077, China
- Department of Chemical Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR, 999077, China
| | - Vivian Wei Yan Wu
- School of Biomedical Sciences, Heart and Vascular Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR, 999077, China
| | - Yumeng Han
- School of Biomedical Sciences, Heart and Vascular Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR, 999077, China
| | - Huiling Hong
- School of Biomedical Sciences, Heart and Vascular Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR, 999077, China
| | - Yalan Wu
- School of Biomedical Sciences, Heart and Vascular Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR, 999077, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, 410013, China
| | - Alice Pik Shan Kong
- Department of Medicine & Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR, 999077, China
| | - Kathy O Lui
- Department of Chemical Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR, 999077, China
| | - Xiao Yu Tian
- School of Biomedical Sciences, Heart and Vascular Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR, 999077, China
| |
Collapse
|
6
|
Bréchot N, Rutault A, Marangon I, Germain S. Blood endothelium transition and phenotypic plasticity: A key regulator of integrity/permeability in response to ischemia. Semin Cell Dev Biol 2024; 155:16-22. [PMID: 37479554 DOI: 10.1016/j.semcdb.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/23/2023]
Abstract
In the human body, the 1013 blood endothelial cells (ECs) which cover a surface of 500-700 m2 (Mai et al., 2013) are key players of tissue homeostasis, remodeling and regeneration. Blood vessel ECs play a major role in the regulation of metabolic and gaz exchanges, cell trafficking, blood coagulation, vascular tone, blood flow and fluid extravasation (also referred to as blood vascular permeability). ECs are heterogeneous in various capillary beds and have the exquisite capacity to cope with environmental changes by regulating their gene expression. Ischemia has major detrimental effects on the endothelium and ischemia-induced regulation of vascular integrity is of paramount importance for human health, as small amounts of fluid accumulation in the interstitium may be responsible for major effects on organ functions and patients outcome. In this review, we will here focus on the stimuli and the molecular mechanisms that control blood endothelium maintenance and phenotypic plasticity/transition involved in controlling blood capillary leakage that might open new avenues for therapeutic applications.
Collapse
Affiliation(s)
- Nicolas Bréchot
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France; Intensive Care Medicine Department, Université de Paris Cité, Hôpital européen Georges-Pompidou, AP-HP, AP-HP.CUP, 75015 Paris, France.
| | - Alexandre Rutault
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France
| | - Iris Marangon
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France.
| |
Collapse
|
7
|
Zhao M, Lei J, Deng F, Zhao C, Xu T, Ji B, Fu M, Wang X, Sun M, Zhang M, Gao Q. Gestational Hypoxia Impaired Endothelial Nitric Oxide Synthesis Via miR-155-5p/NADPH Oxidase/Reactive Oxygen Species Axis in Male Offspring Vessels. J Am Heart Assoc 2024; 13:e032079. [PMID: 38240225 PMCID: PMC11056123 DOI: 10.1161/jaha.123.032079] [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: 08/03/2023] [Accepted: 12/08/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND Nitric oxide (NO) is the most important vasodilator secreted by vascular endothelial cells, and its abnormal synthesis is involved in the development of cardiovascular disease. The prenatal period is a critical time for development and largely determines lifelong vascular health in offspring. Given the high incidence and severity of gestational hypoxia in mid-late pregnancy, it is urgent to further explore whether it affects the long-term synthesis of NO in offspring vascular endothelial cells. METHODS AND RESULTS Pregnant Sprague-Dawley rats were housed in a normoxic or hypoxic (10.5% O2) chamber from gestation days 10 to 20. The thoracic aortas of fetal and adult male offspring were isolated for experiments. Gestational hypoxia significantly reduces the NO-dependent vasodilation mediated by acetylcholine in both the fetal and adult offspring thoracic aorta rings. Meanwhile, acetylcholine-induced NO synthesis is impaired in vascular endothelial cells from hypoxic offspring thoracic aortas. We demonstrate that gestational hypoxic offspring exhibit a reduced endothelial NO synthesis capacity, primarily due to increased expression of NADPH oxidase 2 and enhanced reactive oxygen species. Additionally, gestational hypoxic offspring show elevated levels of miR-155-5p in vascular endothelial cells, which is associated with increased expression of NADPH oxidase 2 and reactive oxygen species generation, as well as impaired NO synthesis. CONCLUSIONS The present study is the first to demonstrate that gestational hypoxia impairs endothelial NO synthesis via the miR-155-5p/NADPH oxidase 2/reactive oxygen species axis in offspring vessels. These novel findings indicate that the detrimental effects of gestational hypoxia on fetal vascular function can persist into adulthood, providing new insights into the development of vascular diseases.
Collapse
Affiliation(s)
- Meng Zhao
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of ChinaMaternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanShandongChina
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
- Department of Obstetrics and GynecologyThe Third People’s Hospital of Bengbu Affiliated to Bengbu Medical CollegeBengbuAnhui ProvinceChina
| | - Jiahui Lei
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Fengying Deng
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Chenxuan Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Ting Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Bingyu Ji
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Mengyu Fu
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xietong Wang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of ChinaMaternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanShandongChina
| | - Miao Sun
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of ChinaMaternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanShandongChina
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health CommissionShandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao UniversityJinanShandongChina
| | - Meihua Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of ChinaMaternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanShandongChina
| | - Qinqin Gao
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of ChinaMaternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanShandongChina
- Institute for Fetology, The First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
8
|
Chatterjee B, Fatima F, Seth S, Sinha Roy S. Moderate Elevation of Homocysteine Induces Endothelial Dysfunction through Adaptive UPR Activation and Metabolic Rewiring. Cells 2024; 13:214. [PMID: 38334606 PMCID: PMC10854856 DOI: 10.3390/cells13030214] [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: 11/06/2023] [Accepted: 11/25/2023] [Indexed: 02/10/2024] Open
Abstract
Elevation of the intermediate amino acid metabolite Homocysteine (Hcy) causes Hyperhomocysteinemia (HHcy), a metabolic disorder frequently associated with mutations in the methionine-cysteine metabolic cycle as well as with nutritional deficiency and aging. The previous literature suggests that HHcy is a strong risk factor for cardiovascular diseases. Severe HHcy is well-established to correlate with vascular pathologies primarily via endothelial cell death. Though moderate HHcy is more prevalent and associated with an increased risk of cardiovascular abnormalities in later part of life, its precise role in endothelial physiology is largely unknown. In this study, we report that moderate elevation of Hcy causes endothelial dysfunction through impairment of their migration and proliferation. We established that unlike severe elevation of Hcy, moderate HHcy is not associated with suppression of endothelial VEGF/VEGFR transcripts and ROS induction. We further showed that moderate HHcy induces a sub-lethal ER stress that causes defective endothelial migration through abnormal actin cytoskeletal remodeling. We also found that sub-lethal increase in Hcy causes endothelial proliferation defect by suppressing mitochondrial respiration and concomitantly increases glycolysis to compensate the consequential ATP loss and maintain overall energy homeostasis. Finally, analyzing a previously published microarray dataset, we confirmed that these hallmarks of moderate HHcy are conserved in adult endothelial cells as well. Thus, we identified adaptive UPR and metabolic rewiring as two key mechanistic signatures in moderate HHcy-associated endothelial dysfunction. As HHcy is clinically associated with enhanced vascular inflammation and hypercoagulability, identifying these mechanistic pathways may serve as future targets to regulate endothelial function and health.
Collapse
Affiliation(s)
- Barun Chatterjee
- CSIR-Institute of Genomics & Integrative Biology, New Delhi 110025, India; (B.C.); (F.F.); (S.S.)
- Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| | - Fabeha Fatima
- CSIR-Institute of Genomics & Integrative Biology, New Delhi 110025, India; (B.C.); (F.F.); (S.S.)
| | - Surabhi Seth
- CSIR-Institute of Genomics & Integrative Biology, New Delhi 110025, India; (B.C.); (F.F.); (S.S.)
- Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| | - Soumya Sinha Roy
- CSIR-Institute of Genomics & Integrative Biology, New Delhi 110025, India; (B.C.); (F.F.); (S.S.)
- Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| |
Collapse
|
9
|
Marzoog BA. Autophagy Behavior in Endothelial Cell Regeneration. Curr Aging Sci 2024; 17:58-67. [PMID: 37861048 DOI: 10.2174/0118746098260689231002044435] [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: 06/28/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023]
Abstract
Autophagy plays a crucial role in maintaining endothelial cell homeostasis through the turnover of intracellular components during stress conditions in a lysosomal-dependent manner. The regeneration strategy involves several aspects, including autophagy. Autophagy is a catabolic degenerative lysosomal-dependent degradation of intracellular components. Autophagy modifies cellular and subcellular endothelial cell functions, including mitochondria stress, lysosomal stress, and endoplasmic reticulum unfolded protein response. Activation of common signaling pathways of autophagy and regeneration and enhancement of intracellular endothelial cell metabolism serve as the bases for the induction of endothelial regeneration. Endothelial progenitor cells include induced pluripotent stem cells (iPSC), embryonic stem cells, and somatic cells, such as fibroblasts. Future strategies of endothelial cell regeneration involve the induction of autophagy to minimize the metabolic degeneration of the endothelial cells and optimize the regeneration outcomes.
Collapse
Affiliation(s)
- Basheer Abdullah Marzoog
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia
| |
Collapse
|
10
|
Johnson BM, Johnson AM, Heim M, Buckley M, Mortimer B, Berry JL, Sewell-Loftin MK. Biomechanical stimulation promotes blood vessel growth despite VEGFR-2 inhibition. BMC Biol 2023; 21:290. [PMID: 38072992 PMCID: PMC10712065 DOI: 10.1186/s12915-023-01792-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Angiogenesis, or the growth of new vasculature from existing blood vessels, is widely considered a primary hallmark of cancer progression. When a tumor is small, diffusion is sufficient to receive essential nutrients; however, as the tumor grows, a vascular supply is needed to deliver oxygen and nutrients into the increasing mass. Several anti-angiogenic cancer therapies target VEGF and the receptor VEGFR-2, which are major promoters of blood vessel development. Unfortunately, many of these cancer treatments fail to completely stop angiogenesis in the tumor microenvironment (TME). Since these therapies focus on the biochemical activation of VEGFR-2 via VEGF ligand binding, we propose that mechanical cues, particularly those found in the TME, may be a source of VEGFR-2 activation that promotes growth of blood vessel networks even in the presence of VEGF and VEGFR-2 inhibitors. RESULTS In this paper, we analyzed phosphorylation patterns of VEGFR-2, particularly at Y1054/Y1059 and Y1214, stimulated via either VEGF or biomechanical stimulation in the form of tensile strains. Our results show prolonged and enhanced activation at both Y1054/Y1059 and Y1214 residues when endothelial cells were stimulated with strain, VEGF, or a combination of both. We also analyzed Src expression, which is downstream of VEGFR-2 and can be activated through strain or the presence of VEGF. Finally, we used fibrin gels and microfluidic devices as 3D microtissue models to simulate the TME. We determined that regions of mechanical strain promoted increased vessel growth, even with VEGFR-2 inhibition through SU5416. CONCLUSIONS Overall, understanding both the effects that biomechanical and biochemical stimuli have on VEGFR-2 activation and angiogenesis is an important factor in developing effective anti-angiogenic therapies. This paper shows that VEGFR-2 can be mechanically activated through strain, which likely contributes to increased angiogenesis in the TME. These proof-of-concept studies show that small molecular inhibitors of VEGFR-2 do not fully prevent angiogenesis in 3D TME models when mechanical strains are introduced.
Collapse
Affiliation(s)
- Bronte Miller Johnson
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6th Avenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, USA
| | - Allison McKenzie Johnson
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6th Avenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, USA
| | - Michael Heim
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6th Avenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, USA
| | - Molly Buckley
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6th Avenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, USA
| | - Bryan Mortimer
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Joel L Berry
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6th Avenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6th Avenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
11
|
Li S, Li J, Cheng W, He W, Dai SS. Independent and Interactive Roles of Immunity and Metabolism in Aortic Dissection. Int J Mol Sci 2023; 24:15908. [PMID: 37958896 PMCID: PMC10647240 DOI: 10.3390/ijms242115908] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/22/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Aortic dissection (AD) is a cardiovascular disease that seriously endangers the lives of patients. The mortality rate of this disease is high, and the incidence is increasing annually, but the pathogenesis of AD is complicated. In recent years, an increasing number of studies have shown that immune cell infiltration in the media and adventitia of the aorta is a novel hallmark of AD. These cells contribute to changes in the immune microenvironment, which can affect their own metabolism and that of parenchymal cells in the aortic wall, which are essential factors that induce degeneration and remodeling of the vascular wall and play important roles in the formation and development of AD. Accordingly, this review focuses on the independent and interactive roles of immunity and metabolism in AD to provide further insights into the pathogenesis, novel ideas for diagnosis and new strategies for treatment or early prevention of AD.
Collapse
Affiliation(s)
- Siyu Li
- School of Medicine, Chongqing University, Chongqing 400044, China
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jun Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wei Cheng
- Department of Cardiac Surgery, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wenhui He
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shuang-Shuang Dai
- School of Medicine, Chongqing University, Chongqing 400044, China
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
12
|
Akhter MS, Goodwin JE. Endothelial Dysfunction in Cardiorenal Conditions: Implications of Endothelial Glucocorticoid Receptor-Wnt Signaling. Int J Mol Sci 2023; 24:14261. [PMID: 37762564 PMCID: PMC10531724 DOI: 10.3390/ijms241814261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
The endothelium constitutes the innermost lining of the blood vessels and controls blood fluidity, vessel permeability, platelet aggregation, and vascular tone. Endothelial dysfunction plays a key role in initiating a vascular inflammatory cascade and is the pivotal cause of various devastating diseases in multiple organs including the heart, lung, kidney, and brain. Glucocorticoids have traditionally been used to combat vascular inflammation. Endothelial cells express glucocorticoid receptors (GRs), and recent studies have demonstrated that endothelial GR negatively regulates vascular inflammation in different pathological conditions such as sepsis, diabetes, and atherosclerosis. Mechanistically, the anti-inflammatory effects of GR are mediated, in part, through the suppression of Wnt signaling. Moreover, GR modulates the fatty acid oxidation (FAO) pathway in endothelial cells and hence can influence FAO-mediated fibrosis in several organs including the kidneys. This review summarizes the relationship between GR and Wnt signaling in endothelial cells and the effects of the Wnt pathway in different cardiac and renal diseases. Available data suggest that GR plays a significant role in restoring endothelial integrity, and research on endothelial GR-Wnt interactions could facilitate the development of novel therapies for many cardiorenal conditions.
Collapse
Affiliation(s)
- Mohammad Shohel Akhter
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06511, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Julie Elizabeth Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06511, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06511, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06511, USA
| |
Collapse
|
13
|
Braczko A, Harasim G, Kawecka A, Walczak I, Kapusta M, Narajczyk M, Stawarska K, Smoleński RT, Kutryb-Zając B. Blocking cholesterol formation and turnover improves cellular and mitochondria function in murine heart microvascular endothelial cells and cardiomyocytes. Front Physiol 2023; 14:1216267. [PMID: 37745244 PMCID: PMC10512729 DOI: 10.3389/fphys.2023.1216267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023] Open
Abstract
Background: Statins and proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) are cornerstones of therapy to prevent cardiovascular disease, acting by lowering lipid concentrations and only partially identified pleiotropic effects. This study aimed to analyze impacts of atorvastatin and synthetic peptide PCSK9i on bioenergetics and function of microvascular endothelial cells and cardiomyocytes. Methods: Mitochondrial function and abundance as well as intracellular nucleotides, membrane potential, cytoskeleton structure, and cell proliferation rate were evaluated in mouse heart microvascular endothelial cells (H5V) and cardiomyocytes (HL-1) under normal and hypoxia-mimicking conditions (CoCl2 exposure). Results: In normal conditions PCSK9i, unlike atorvastatin, enhanced mitochondrial respiratory parameters, increased nucleotide levels, prevented actin cytoskeleton disturbances and stimulated endothelial cell proliferation. Under hypoxia-mimicking conditions both atorvastatin and PCSK9i improved the mitochondrial respiration and membrane potential in both cell types. Conclusion: This study demonstrated that both treatments benefited the endothelial cell and cardiomyocyte bioenergetics, but the effects of PCSK9i were superior.
Collapse
Affiliation(s)
- Alicja Braczko
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Gabriela Harasim
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Ada Kawecka
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Iga Walczak
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | | | | | - Klaudia Stawarska
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | | | | |
Collapse
|
14
|
Le NT. Metabolic regulation of endothelial senescence. Front Cardiovasc Med 2023; 10:1232681. [PMID: 37649668 PMCID: PMC10464912 DOI: 10.3389/fcvm.2023.1232681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/18/2023] [Indexed: 09/01/2023] Open
Abstract
Endothelial cell (EC) senescence is increasingly recognized as a significant contributor to the development of vascular dysfunction and age-related disorders and diseases, including cancer and cardiovascular diseases (CVD). The regulation of cellular senescence is known to be influenced by cellular metabolism. While extensive research has been conducted on the metabolic regulation of senescence in other cells such as cancer cells and fibroblasts, our understanding of the metabolic regulation of EC senescence remains limited. The specific metabolic changes that drive EC senescence are yet to be fully elucidated. The objective of this review is to provide an overview of the intricate interplay between cellular metabolism and senescence, with a particular emphasis on recent advancements in understanding the metabolic changes preceding cellular senescence. I will summarize the current knowledge on the metabolic regulation of EC senescence, aiming to offer insights into the underlying mechanisms and future research directions.
Collapse
Affiliation(s)
- Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
15
|
Marzoog BA. Endothelial cell autophagy in the context of disease development. Anat Cell Biol 2023; 56:16-24. [PMID: 36267005 PMCID: PMC9989784 DOI: 10.5115/acb.22.098] [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: 05/11/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 11/27/2022] Open
Abstract
Endothelial cells (EC) are the anatomical boundaries between the intravascular and extravascular space. Damage to ECs is catastrophic and induces endothelial cell dysfunction. The pathogenesis is multifactorial and involves dysregulation in the signaling pathways, membrane lipids ratio disturbance, cell-cell adhesion disturbance, unfolded protein response, lysosomal and mitochondrial stress, autophagy dysregulation, and oxidative stress. Autophagy is a lysosomal-dependent turnover of intracellular components. Autophagy was recognized early in the pathogenesis of endothelial dysfunction. Autophagy is a remarkable patho (physiological) process in the cell homeostasis regulation including EC. Regulation of autophagy rate is disease-dependent and impaired with aging. Up-regulation of autophagy induces endothelial cell regeneration/differentiation and improves the function of impaired ones. The paper scrutinizes the molecular mechanisms and triggers of EC dysregulation and current perspectives for future therapeutic strategies by autophagy targeting.
Collapse
|
16
|
Akhter MS, Kubra KT, Barabutis N. Protective effects of GHRH antagonists against hydrogen peroxide-induced lung endothelial barrier disruption. Endocrine 2023; 79:587-592. [PMID: 36261700 PMCID: PMC9581763 DOI: 10.1007/s12020-022-03226-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/04/2022] [Indexed: 12/02/2022]
Abstract
PURPOSE Growth hormone-releasing hormone (GHRH) is a hypothalamic hormone, which regulates growth hormone release from the anterior pituitary gland. GHRH antagonists (GHRHAnt) are anticancer agents, which also exert robust anti-inflammatory activities in malignancies. GHRHAnt exhibit anti-oxidative and anti-inflammatory effects in vascular endothelial cells, indicating their potential use against disorders related to barrier dysfunction (e.g. sepsis). Herein, we aim to investigate the effects of GHRHAnt against lung endothelial hyperpermeability. METHODS The in vitro effects of GHRHAnt in H2O2-induced endothelial barrier dysfunction were investigated in bovine pulmonary artery endothelial cells (BPAEC). Electric cell-substrate impedance sensing (ECIS) was utilized to measure transendothelial resistance, an indicator of barrier function. RESULTS Our results demonstrate that GHRHAnt protect against H2O2-induced endothelial barrier disruption via P53 and cofilin modulation. Both proteins are crucial modulators of vascular integrity. Moreover, GHRHAnt prevent H2O2 - induced decrease in transendothelial resistance. CONCLUSIONS GHRHAnt represent a promising therapeutic intervention towards diseases related to lung endothelial hyperpermeability, such as acute respiratory distress syndrome - related or not to COVID-19 - and sepsis. Targeted medicine for those potentially lethal disorders does not exist.
Collapse
Affiliation(s)
- Mohammad S Akhter
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, 71201, USA
| | - Khadeja-Tul Kubra
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, 71201, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, 71201, USA.
| |
Collapse
|
17
|
Proteins Adsorbed during Intraoperative Hemoadsorption and Their In Vitro Effects on Endothelium. Healthcare (Basel) 2023; 11:healthcare11030310. [PMID: 36766885 PMCID: PMC9914797 DOI: 10.3390/healthcare11030310] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
(1) Background: Hemoadsorption is a method of blood purification with a wide spectrum of indications. Pre-emptive use of hemoadsorption in patients undergoing heart surgery with cardiopulmonary bypass is considered to reduce the risk of postoperative systemic inflammatory response syndrome. The current study aimed to identify the spectrum of blood proteins adsorbed on the polymer matrix of the CytoSorb hemoadsorption system and to investigate their influence on cultured endothelial cells in vitro. (2) Methods: Adsorbers used for intraoperative hemoadsorption were obtained from patients undergoing on-pump valve surgery in acute endocarditis. Proteins were extracted from the adsorbers, purified, identified with mass-spectrometry and applied to cultured human aortic endothelial cells. (3) Results: A broad range of blood proteins were identified in the material eluted from the CytoSorb adsorber. When added to cultured ECs, these protein extracts caused severe reduction in cell viability and migration. After 24 h exposure, transcriptional changes with up-regulation of multiple metabolic regulators were observed and verified on the protein level. Genes responsible for control of mitosis were significantly down-regulated. (4) Conclusions: In summary, our data reveal that intraoperative hemoadsorption allows broad spectrum removal of a wide range of molecules eliciting endothelial damage.
Collapse
|
18
|
Barabutis N, Akhter MS, Kubra KT, Jackson K. Growth Hormone-Releasing Hormone in Endothelial Inflammation. Endocrinology 2022; 164:6887354. [PMID: 36503995 PMCID: PMC9923806 DOI: 10.1210/endocr/bqac209] [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: 06/08/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
The discovery of hypothalamic hormones propelled exciting advances in pharmacotherapy and improved life quality worldwide. Growth hormone-releasing hormone (GHRH) is a crucial element in homeostasis maintenance, and regulates the release of growth hormone from the anterior pituitary gland. Accumulating evidence suggests that this neuropeptide can also promote malignancies, as well as inflammation. Our review is focused on the role of that 44 - amino acid peptide (GHRH) and its antagonists in inflammation and vascular function, summarizing recent findings in the corresponding field. Preclinical studies demonstrate the protective role of GHRH antagonists against endothelial barrier dysfunction, suggesting that the development of those peptides may lead to new therapies against pathologies related to vascular remodeling (eg, sepsis, acute respiratory distress syndrome). Targeted therapies for those diseases do not exist.
Collapse
Affiliation(s)
- Nektarios Barabutis
- Correspondence: Nektarios Barabutis, MSc, PhD, School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Dr, Monroe, LA 71201, USA.
| | | | - Khadeja-Tul Kubra
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Keith Jackson
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| |
Collapse
|
19
|
Atallah R, Olschewski A, Heinemann A. Succinate at the Crossroad of Metabolism and Angiogenesis: Roles of SDH, HIF1α and SUCNR1. Biomedicines 2022; 10:3089. [PMID: 36551845 PMCID: PMC9775124 DOI: 10.3390/biomedicines10123089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Angiogenesis is an essential process by which new blood vessels develop from existing ones. While adequate angiogenesis is a physiological process during, for example, tissue repair, insufficient and excessive angiogenesis stands on the pathological side. Fine balance between pro- and anti-angiogenic factors in the tissue environment regulates angiogenesis. Identification of these factors and how they function is a pressing topic to develop angiogenesis-targeted therapeutics. During the last decade, exciting data highlighted non-metabolic functions of intermediates of the mitochondrial Krebs cycle including succinate. Among these functions is the contribution of succinate to angiogenesis in various contexts and through different mechanisms. As the concept of targeting metabolism to treat a wide range of diseases is rising, in this review we summarize the mechanisms by which succinate regulates angiogenesis in normal and pathological settings. Gaining a comprehensive insight into how this metabolite functions as an angiogenic signal will provide a useful approach to understand diseases with aberrant or excessive angiogenic background, and may provide strategies to tackle them.
Collapse
Affiliation(s)
- Reham Atallah
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Akos Heinemann
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| |
Collapse
|
20
|
Hanser S, Mphekgwana PM, Moraba MM, Erasmus L, van Staden M. Increased endothelial biomarkers are associated with HIV antiretroviral therapy and C-reactive protein among a African rural population in Limpopo Province, South Africa. Front Public Health 2022; 10:980754. [PMID: 36407976 PMCID: PMC9672841 DOI: 10.3389/fpubh.2022.980754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
In Sub-Saharan Africa (SSA) endothelial dysfunction (ED) and chronic inflammation in the HIV-positive adults population who are on highly active antiretroviral therapy (HAART) are not fully explored. We determined the effect of HAART on chronic inflammation and ED among HAART-exposed adults in a rural setting. Weight and height were measured to quantify the body mass index (BMI). Lipid and Glucose levels were determined. C-reactive protein (CRP), L-selectin, soluble intercellular adhesion molecule (sICAM-1), and soluble vascular cell adhesion molecule (sVCAM-1) in serum samples were tested. The majority of the HAART-exposed group were on treatment for <5 years. Soluble intercellular adhesion molecules, sVCAM-1, L-selectin and CRP were elevated in the HIV-infected groups as compared to the control group. The multivariate analysis showed that HIV infection (HAART-naïve) associated with increased sICAM-1 (β = 0.350; 95% CI: 0.035-0.664, p = 0.029) and L-selectin (β = 0.236; 95% CI: 0.038-0.434, p = 0.019) but not sVCAM-1 (β = 0.009; 95% CI: 0.252-0.270, p = 0.468). The HAART-exposed group is associated with sVCAM-1 (β = 0.250; 95% CI: 0.015-0.486, p = 0.037) but not with sICAM-1- (β = 0.253; 95% CI: -0.083-0.590, p = 0.14) and L-selectin (β = 0.119; 95% CI: -0.016-0.253, p = 0.084). sVCAM-1 was associated with decreased alcohol consumption (β = -0.245; 95% CI: -0.469-0.021, p = 0.032) while L-selectin was associated with decreased total cholesterol (β = -0.061; 95% CI: -0.124-0.002, p = 0.05) and increased CRP (β = 0.015; 95% CI: 0.009-0.022, p < 0.001). Increased endothelial biomarkers were associated with HIV disease and HAART in a rural black adult population of African descent after controlling for CVD risk factors. Inflammation (as measured with CRP) may play an important role in endothelial activation. Further studies are needed to explore the association between endothelial dysfunction and inflammation especially among the HIV-positive population on HAART in similar settings.
Collapse
Affiliation(s)
- Sidney Hanser
- Department of Physiology and Environmental Health, University of Limpopo, Polokwane, South Africa,*Correspondence: Sidney Hanser
| | | | | | - Lourens Erasmus
- Department of Physiology and Environmental Health, University of Limpopo, Polokwane, South Africa
| | - Marlise van Staden
- Department of Physiology and Environmental Health, University of Limpopo, Polokwane, South Africa
| |
Collapse
|
21
|
Disturbance of Fatty Acid Metabolism Promoted Vascular Endothelial Cell Senescence via Acetyl-CoA-Induced Protein Acetylation Modification. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1198607. [PMID: 35993026 PMCID: PMC9385365 DOI: 10.1155/2022/1198607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/27/2022] [Accepted: 07/01/2022] [Indexed: 11/18/2022]
Abstract
Endothelial cell senescence is the main risk factor contributing to vascular dysfunction and the progression of aging-related cardiovascular diseases. However, the relationship between endothelial cell metabolism and endothelial senescence remains unclear. The present study provides novel insight into fatty acid metabolism in the regulation of endothelial senescence. In the replicative senescence model and H2O2-induced premature senescence model of primary cultured human umbilical vein endothelial cells (HUVECs), fatty acid oxidation (FAO) was suppressed and fatty acid profile was disturbed, accompanied by downregulation of proteins associated with fatty acid uptake and mitochondrial entry, in particular the FAO rate-limiting enzyme carnitine palmitoyl transferase 1A (CPT1A). Impairment of fatty acid metabolism by silencing CPT1A or CPT1A inhibitor etomoxir facilitated the development of endothelial senescence, as implied by the increase of p53, p21, and senescence-associated β-galactosidase, as well as the decrease of EdU-positive proliferating cells. In the contrary, rescue of FAO by overexpression of CPT1A or supplement of short chain fatty acids (SCFAs) acetate and propionate ameliorated endothelial senescence. In vivo, treatment of acetate for 4 weeks lowered the blood pressure and alleviated the senescence-related phenotypes in aortas of Ang II-infused mice. Mechanistically, fatty acid metabolism regulates endothelial senescence via acetyl-coenzyme A (acetyl-CoA), as implied by the observations that suppression of acetyl-CoA production using the inhibitor of ATP citrate lyase NDI-091143 accelerated senescence of HUVECs and that supplementation of acetyl-CoA prevented H2O2-induced endothelial senescence. Deficiency of acetyl-CoA resulted in alteration of acetylated protein profiles which are associated with cell metabolism and cell cycle. These findings thus suggest that improvement of fatty acid metabolism might ameliorate endothelial senescence-associated cardiovascular diseases.
Collapse
|
22
|
Zhou Q, Guo B, Chen D, Yao H, Liang X, Xin J, Shi D, Ren K, Yang H, Jiang J, Li J. Dynamic Alterations of Metabolites Revealed the Vascularization Progression of Bioengineered Liver. Biotechnol Bioeng 2022; 119:2857-2867. [PMID: 35864592 DOI: 10.1002/bit.28189] [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: 09/21/2021] [Revised: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 11/08/2022]
Abstract
Vascularization is a critical but challenging process in developing functional bioengineered liver with the decellularized liver scaffolds (DLSs), and the process is accompanied by cell-specific metabolic alterations. To elucidate the dynamic alterations of metabolites during vascularization, rat DLSs were vascularized with human umbilical vein endothelial cells, and a liquid chromatography mass spectrometry-based metabolomics was performed on culture supernatants collected at 0, 1, 3, 7, 14 and 21 days. Overall, 1698 peak pairs or metabolites were detected in the culture supernatants, with 309 metabolites being positively identified. The orthogonal partial least-squares discriminant analysis and functional enrichment analysis revealed three phases that could be clearly discriminated, including phase D1 (cell proliferation and migration), phase D3D7 (vascular lumen formation), and phase D14D21 (functional endothelial barrier formation). Seventy-two common differentially abundant metabolites of known identity were detected in these three phases when compared to day 0. Of these metabolites, a high level of beta-Alanine indicated a better degree of vascularization, and 14 days of in-vitro dynamic culture is required to develop a functionalized vascular structure. These results enriched our understanding of the metabolic mechanism of DLS vascularization, and indicated that beta-Alanine could function as a potential predictor of the patency of vascularized bioengineered livers. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Qian Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Beibei Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Deying Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Heng Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Xi Liang
- Precision Medicine Center of Taizhou Central Hospital, Taizhou University Medical School, Taizhou, China
| | - Jiaojiao Xin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China.,Precision Medicine Center of Taizhou Central Hospital, Taizhou University Medical School, Taizhou, China
| | - Dongyan Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China.,Precision Medicine Center of Taizhou Central Hospital, Taizhou University Medical School, Taizhou, China
| | - Keke Ren
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Hui Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Jing Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China.,Precision Medicine Center of Taizhou Central Hospital, Taizhou University Medical School, Taizhou, China
| | - Jun Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China.,Precision Medicine Center of Taizhou Central Hospital, Taizhou University Medical School, Taizhou, China
| |
Collapse
|
23
|
CoCl2-Mimicked Endothelial Cell Hypoxia Induces Nucleotide Depletion and Functional Impairment That Is Reversed by Nucleotide Precursors. Biomedicines 2022; 10:biomedicines10071540. [PMID: 35884844 PMCID: PMC9313011 DOI: 10.3390/biomedicines10071540] [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: 05/26/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic hypoxia drives vascular dysfunction by various mechanisms, including changes in mitochondrial respiration. Although endothelial cells (ECs) rely predominantly on glycolysis, hypoxia is known to alter oxidative phosphorylation, promote oxidative stress and induce dysfunction in ECs. Our work aimed to analyze the effects of prolonged treatment with hypoxia-mimetic agent CoCl2 on intracellular nucleotide concentration, extracellular nucleotide breakdown, mitochondrial function, and nitric oxide (NO) production in microvascular ECs. Moreover, we investigated how nucleotide precursor supplementation and adenosine deaminase inhibition protected against CoCl2-mediated disturbances. Mouse (H5V) and human (HMEC-1) microvascular ECs were exposed to CoCl2-mimicked hypoxia for 24 h in the presence of nucleotide precursors: adenine and ribose, and adenosine deaminase inhibitor, 2′deoxycoformycin. CoCl2 treatment decreased NO production by ECs, depleted intracellular ATP concentration, and increased extracellular nucleotide and adenosine catabolism in both H5V and HMEC-1 cell lines. Diminished intracellular ATP level was the effect of disturbed mitochondrial phosphorylation, while nucleotide precursors effectively restored the ATP pool via the salvage pathway and improved endothelial function under CoCl2 treatment. Endothelial protective effects of adenine and ribose were further enhanced by adenosine deaminase inhibition, that increased adenosine concentration. This work points to a novel strategy for protection of hypoxic ECs by replenishing the adenine nucleotide pool and promoting adenosine signaling.
Collapse
|
24
|
Wu WP, Zhou MY, Liu DL, Min X, Shao T, Xu ZY, Jing X, Cai MY, Xu S, Liang X, Mo M, Liu X, Xiong XD. circGNAQ, a circular RNA enriched in vascular endothelium, inhibits endothelial cell senescence and atherosclerosis progression. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:374-387. [PMID: 34552819 PMCID: PMC8426466 DOI: 10.1016/j.omtn.2021.07.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/30/2021] [Indexed: 11/19/2022]
Abstract
Endothelial cell senescence is one of the most important causes of vascular dysfunction and atherosclerosis. Circular RNAs (circRNAs) are endogenous RNA molecules with covalently closed-loop structures, which have been reported to be abnormally expressed in many human diseases. However, the potential role of circRNAs in endothelial cell senescence and atherosclerosis remains largely unknown. Here, we compared the expression patterns of circRNAs in young and senescent human endothelial cells with RNA sequencing. Among the differentially expressed circRNAs, circGNAQ, a circRNA enriched in vascular endothelium, was significantly downregulated in senescent endothelial cells. circGNAQ silencing triggered endothelial cell senescence, as determined by a rise in senescence-associated β-galactosidase activity, reduced cell proliferation, and suppressed angiogenesis; circGNAQ overexpression showed the opposite effects. Mechanistic studies revealed that circGNAQ acted as an endogenous miR-146a-5p sponge to increase the expression of its target gene PLK2 by decoying the miR-146a-5p, thereby delaying endothelial cell senescence. In vivo studies showed that circGNAQ overexpression in the endothelium inhibited endothelial cell senescence and atherosclerosis progression. These results suggest that circGNAQ plays critical roles in endothelial cell senescence and consequently the pathogenesis of atherosclerosis, implying that the management of circGNAQ provides a potential therapeutic approach for limiting the progression of atherosclerosis.
Collapse
Affiliation(s)
- Wei-peng Wu
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Meng-yuan Zhou
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Dong-liang Liu
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Xue Min
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Tong Shao
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Zi-yang Xu
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Xia Jing
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Meng-yun Cai
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Shun Xu
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Xin Liang
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Miaohua Mo
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Xinguang Liu
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
- Institute of Biochemistry & Molecular Biology, Guangdong Medical University, Zhanjiang 524023, P.R. China
| | - Xing-dong Xiong
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China
- Institute of Biochemistry & Molecular Biology, Guangdong Medical University, Zhanjiang 524023, P.R. China
- Corresponding author: Prof. Xing-dong Xiong, Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, P.R. China.
| |
Collapse
|
25
|
Rodríguez CM, Velásquez-Berrío M, Rúa C, Viana M, Abrahams VM, Cadavid AP, Alvarez AM. Antiphospholipid Antibodies From Women With Pregnancy Morbidity and Vascular Thrombosis Induce Endothelial Mitochondrial Dysfunction, mTOR Activation, and Autophagy. Front Physiol 2021; 12:706743. [PMID: 34912234 PMCID: PMC8667788 DOI: 10.3389/fphys.2021.706743] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 10/18/2021] [Indexed: 01/09/2023] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease characterized by thrombosis and pregnancy morbidity (PM) obstetric events together with persistent high titers of circulating antiphospholipid antibodies (aPL). Several mechanisms that explain the development of thrombosis and PM in APS include the association of aPL with alterations in the coagulation cascade and inflammatory events. Other mechanisms disturbing cellular homeostases, such as mitochondrial dysfunction, autophagy, and cell proliferation, have been described in other autoimmune diseases. Therefore, the objective of this study was to investigate the impact of aPL from different patient populations on endothelial cell mitochondrial function, activation of the mammalian target of rapamycin (mTOR) and autophagy pathways, and cellular growth. Using an in vitro model, human umbilical vein endothelial cells (HUVECs) were treated with polyclonal immunoglobulin G (IgG) purified from the serum of women with both PM and vascular thrombosis (PM/VT), with VT only (VT), or with PM and non-criteria aPL (seronegative-obstetric APS, SN-OAPS). We included IgG from women with PM without aPL (PM/aPL-) and healthy women with previous uncomplicated pregnancies (normal human serum, NHS) as control groups. Mitochondrial function, mTOR activation, autophagy, and cell proliferation were evaluated by Western blotting, flow cytometry, and functional assays. IgG from women with PM/VT increased HUVEC mitochondrial hyperpolarization and activation of the mTOR and autophagic pathways, while IgG from patients with VT induced endothelial autophagy and cell proliferation in the absence of elevated mTOR activity or mitochondrial dysfunction. IgG from the SN-OAPS patient group had no effect on any of these HUVEC responses. In conclusion, aPL from women with PM and vascular events induce cellular stress evidenced by mitochondrial hyperpolarization and increased activation of the mTOR and autophagic pathways which may play a role in the pathogenesis of obstetric APS.
Collapse
Affiliation(s)
- Carlos M. Rodríguez
- Grupo Reproducción, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Manuela Velásquez-Berrío
- Grupo Reproducción, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Carolina Rúa
- Grupo de Investigación en Trombosis, Facultad de Medicina, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Marta Viana
- Grupo de Metabolismo y Función Vascular, Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Red Iberoamericana de Alteraciones Vasculares Asociadas a Transtornos del Embarazo (RIVATREM), Chillán, Chile
| | - Vikki M. Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States
| | - Angela P. Cadavid
- Grupo Reproducción, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia (UdeA), Medellín, Colombia
- Red Iberoamericana de Alteraciones Vasculares Asociadas a Transtornos del Embarazo (RIVATREM), Chillán, Chile
| | - Angela M. Alvarez
- Grupo Reproducción, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia (UdeA), Medellín, Colombia
| |
Collapse
|
26
|
The Blood-Brain Barrier: Much More Than a Selective Access to the Brain. Neurotox Res 2021; 39:2154-2174. [PMID: 34677787 DOI: 10.1007/s12640-021-00431-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/30/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022]
Abstract
The blood-brain barrier is a dynamic structure, collectively referred to as the neurovascular unit. It is responsible for the exchange of blood, oxygen, ions, and other molecules between the peripheral circulation and the brain compartment. It is the main entrance to the central nervous system and as such critical for the maintenance of its homeostasis. Dysfunction of the blood-brain barrier is a characteristic of several neurovascular pathologies. Moreover, physiological changes, environmental factors, nutritional habits, and psychological stress can modulate the tightness of the barrier. In this contribution, we summarize our current understanding of structure and function of this important component of the brain. We also describe the neurological deficits associated with its damage. A special emphasis is placed in the effect of the exposure to xenobiotics and pollutants in the permeability of the barrier. Finally, current protective strategies as well as the culture models to study this fascinating structure are discussed.
Collapse
|
27
|
Ullah K, Wu R. Hypoxia-Inducible Factor Regulates Endothelial Metabolism in Cardiovascular Disease. Front Physiol 2021; 12:670653. [PMID: 34290616 PMCID: PMC8287728 DOI: 10.3389/fphys.2021.670653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/13/2021] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells (ECs) form a physical barrier between the lumens and vascular walls of arteries, veins, capillaries, and lymph vessels; thus, they regulate the extravasation of nutrients and oxygen from the circulation into the perivascular space and participate in mechanisms that maintain cardiovascular homeostasis and promote tissue growth and repair. Notably, their role in tissue repair is facilitated, at least in part, by their dependence on glycolysis for energy production, which enables them to resist hypoxic damage and promote angiogenesis in ischemic regions. ECs are also equipped with a network of oxygen-sensitive molecules that collectively activate the response to hypoxic injury, and the master regulators of the hypoxia response pathway are hypoxia-inducible factors (HIFs). HIFs reinforce the glycolytic dependence of ECs under hypoxic conditions, but whether HIF activity attenuates or exacerbates the progression and severity of cardiovascular dysfunction varies depending on the disease setting. This review summarizes how HIF regulates the metabolic and angiogenic activity of ECs under both normal and hypoxic conditions and in a variety of diseases that are associated with cardiovascular complications.
Collapse
Affiliation(s)
- Karim Ullah
- Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Rongxue Wu
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
28
|
PSMA Expression in 122 Treatment Naive Glioma Patients Related to Tumor Metabolism in 11C-Methionine PET and Survival. J Pers Med 2021; 11:jpm11070624. [PMID: 34209106 PMCID: PMC8305688 DOI: 10.3390/jpm11070624] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/20/2021] [Accepted: 06/24/2021] [Indexed: 12/18/2022] Open
Abstract
Apart from its expression in benign and malignant prostate tissue, prostate specific membrane antigen (PSMA) was shown to be expressed specifically in the neovasculature of solid tumors. For gliomas only little information exists. Therefore, we aimed to correlate PSMA expression in gliomas to tumor metabolism by L-[S-methyl-11C]methionine (MET) PET and survival. Therefore, immunohistochemical staining (IHC) for isocitrate dehydrogenase 1-R132H (IDH1-R132H) mutation and PSMA expression was performed on the paraffin embedded tissue samples of 122 treatment-naive glioma patients. The IHC results were then related to the pre-therapeutic semiquantitative MET PET data and patients' survival. Vascular PSMA expression was observed in 26 of 122 samples and was rather specific for high-grade gliomas ([HGG] 81% of glioblastoma multiforme, 10% of WHO grade III and just 2% of grade II gliomas). Significantly higher amounts of gliomas without verifiable IDH1-R132H mutation showed vascular PSMA expression. Significantly shorter median survival times were seen for patients with vascular PSMA staining in all tumors as well as HGG only. Additionally, significantly higher numbers of PSMA staining vessels were found in tumors with high amino acid metabolic rates. Vascular PSMA expression in gliomas was seen as a high-grade specific feature associated with elevated amino acid metabolism and short survival.
Collapse
|
29
|
Danoy M, Jellali R, Tauran Y, Bruce J, Leduc M, Gilard F, Gakière B, Scheidecker B, Kido T, Miyajima A, Soncin F, Sakai Y, Leclerc E. Characterization of the proteome and metabolome of human liver sinusoidal endothelial-like cells derived from induced pluripotent stem cells. Differentiation 2021; 120:28-35. [PMID: 34229994 DOI: 10.1016/j.diff.2021.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/11/2021] [Accepted: 06/27/2021] [Indexed: 01/22/2023]
Abstract
The liver is a complex organ composed of several cell types organized hierarchically. Among these, liver sinusoidal endothelial cells (LSECs) are specialized vascular cells known to interact with hepatocytes and hepatic stellate cells (HSCs), and to be involved in the regulation of important hepatic processes in healthy and pathological situations. Protocols for the differentiation of LSECs from human induced pluripotent stem cells, hiPSCs, have been proposed and in-depth analysis by transcriptomic profiling of those cells has been performed. In the present work, an extended analysis of those cells in terms of proteome and metabolome has been implemented. The proteomic analysis confirmed the expression of important endothelial markers and pathways. Among them, the expression of patterns typical of LSECs such as PECAM1, VWF, LYVE1, STAB1 (endothelial markers), CDH13, CDH5, CLDN5, ICAM1, MCAM-CD146, ICAM2, ESAM (endothelial cytoskeleton), NOSTRIN, NOS3 (Nitric Oxide endothelial ROS), ESM1, ENG, MMRN2, THBS1, ANGPT2 (angiogenesis), CD93, MRC1 (mannose receptor), CLEC14A (C-type lectin), CD40 (antigen), and ERG (transcription factor) was highlighted. Besides, the pathway analysis revealed the enrichment of the endocytosis, Toll-like receptor, Nod-like receptor, Wnt, Apelin, VEGF, cGMP-PCK, and PPAR related signaling pathways. Other important pathways such as vasopressin regulated water reabsorption, fluid shear stress, relaxin signaling, and renin secretion were also highlighted. At confluence, the metabolome profile appeared consistent with quiescent endothelial cell patterns. The integration of both proteome and metabolome datasets revealed a switch from fatty acid synthesis in undifferentiated hiPSCs to a fatty oxidation in LSECs and activation of the pentose phosphate pathway and polyamine metabolism in hiPSCs-derived LSECs. In conclusion, the comparison between the signature of LSECs differentiated following the protocol described in this work, and data found in the literature confirmed the particular relevance of these cells for future in vitro applications.
Collapse
Affiliation(s)
- Mathieu Danoy
- CNRS UMI 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Rachid Jellali
- Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu CS 60319, 60203, Compiègne Cedex, France
| | - Yannick Tauran
- Univ Lyon, Université Claude Bernard Lyon 1, Laboratoire des Multimatériaux et Interfaces, UMR CNRS 5615, F-69622, Villeurbanne, France
| | - Johanna Bruce
- Plateforme protéomique 3P5, Université de Paris, Institut Cochin, INSERM, CNRS, F-75014, France
| | - Marjorie Leduc
- Plateforme protéomique 3P5, Université de Paris, Institut Cochin, INSERM, CNRS, F-75014, France
| | - Françoise Gilard
- Plateforme Métabolisme Métabolome, Institute of Plant Sciences Paris-Saclay (IPS2), CNRS, INRA, Univ. Paris-Sud, Univ. Evry, Univ. Paris-Diderot, Univ. Paris Saclay, Bâtiment 630 Rue Noetzlin, 91192, Gif-sur-Yvette cedex, France
| | - Bertrand Gakière
- Plateforme Métabolisme Métabolome, Institute of Plant Sciences Paris-Saclay (IPS2), CNRS, INRA, Univ. Paris-Sud, Univ. Evry, Univ. Paris-Diderot, Univ. Paris Saclay, Bâtiment 630 Rue Noetzlin, 91192, Gif-sur-Yvette cedex, France
| | - Benedikt Scheidecker
- CNRS UMI 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Taketomo Kido
- Laboratory of Stem Cell Therapy, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Atsushi Miyajima
- Laboratory of Stem Cell Therapy, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Fabrice Soncin
- CNRS UMI 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; CNRS/IIS/Centre Oscar Lambret/Lille University SMMiL-E Project, CNRS Délégation Nord-Pas de Calais et Picardie, 2 rue de Canonniers, Lille, 59046, France
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Eric Leclerc
- CNRS UMI 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu CS 60319, 60203, Compiègne Cedex, France.
| |
Collapse
|
30
|
Marottoli FM, Trevino TN, Geng X, Arbieva Z, Kanabar P, Maienschein-Cline M, Lee JC, Lutz SE, Tai LM. Autocrine Effects of Brain Endothelial Cell-Produced Human Apolipoprotein E on Metabolism and Inflammation in vitro. Front Cell Dev Biol 2021; 9:668296. [PMID: 34178992 PMCID: PMC8225247 DOI: 10.3389/fcell.2021.668296] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/11/2021] [Indexed: 11/13/2022] Open
Abstract
Reports of APOE4-associated neurovascular dysfunction during aging and in neurodegenerative disorders has led to ongoing research to identify underlying mechanisms. In this study, we focused on whether the APOE genotype of brain endothelial cells modulates their own phenotype. We utilized a modified primary mouse brain endothelial cell isolation protocol that enabled us to perform experiments without subculture. Through initial characterization we found, that compared to APOE3, APOE4 brain endothelial cells produce less apolipoprotein E (apoE) and have altered metabolic and inflammatory gene expression profiles. Further analysis revealed APOE4 brain endothelial cultures have higher preference for oxidative phosphorylation over glycolysis and, accordingly, higher markers of mitochondrial activity. Mitochondrial activity generates reactive oxygen species, and, with APOE4, there were higher mitochondrial superoxide levels, lower levels of antioxidants related to heme and glutathione and higher markers/outcomes of oxidative damage to proteins and lipids. In parallel, or resulting from reactive oxygen species, there was greater inflammation in APOE4 brain endothelial cells including higher chemokine levels and immune cell adhesion under basal conditions and after low-dose lipopolysaccharide (LPS) treatment. In addition, paracellular permeability was higher in APOE4 brain endothelial cells in basal conditions and after high-dose LPS treatment. Finally, we found that a nuclear receptor Rev-Erb agonist, SR9009, improved functional metabolic markers, lowered inflammation and modulated paracellular permeability at baseline and following LPS treatment in APOE4 brain endothelial cells. Together, our data suggest that autocrine signaling of apoE in brain endothelial cells represents a novel cellular mechanism for how APOE regulates neurovascular function.
Collapse
Affiliation(s)
- Felecia M Marottoli
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Troy N Trevino
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Xue Geng
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Zarema Arbieva
- Genome Research Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States
| | - Pinal Kanabar
- Research Informatics Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States
| | - Mark Maienschein-Cline
- Research Informatics Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States
| | - James C Lee
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Sarah E Lutz
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
31
|
Abstract
The endothelium is a crucial regulator of vascular homeostasis by controlling barrier integrity as well acting as an important signal transducer, thereby illustrating that endothelial cells are not inert cells. In the context of atherosclerosis, this barrier function is impaired and endothelial cells become activated, resulting in the upregulation of adhesion molecules, secretion of cytokines and chemokines and internalization of integrins. Finally, this leads to increased vessel permeability, thereby facilitating leukocyte extravasation as well as fostering a pro-inflammatory environment. Additionally, activated endothelial cells can form migrating tip cells and proliferative stalk cells, resulting in the formation of new blood vessels. Emerging evidence has accumulated indicating that cellular metabolism is crucial in fueling these pro-atherosclerotic processes, including neovascularization and inflammation, thereby contributing to plaque progression and altering plaque stability. Therefore, further research is necessary to unravel the complex mechanisms underlying endothelial cell metabolic changes, and exploit this knowledge for finding and developing potential future therapeutic strategies. In this review we discuss the metabolic alterations endothelial cells undergo in the context of inflammation and atherosclerosis and how this relates to changes in endothelial functioning. Finally, we will describe several metabolic targets that are currently being used for therapeutic interventions.
Collapse
|
32
|
Zhou ZY, Wang L, Wang YS, Dou GR. PFKFB3: A Potential Key to Ocular Angiogenesis. Front Cell Dev Biol 2021; 9:628317. [PMID: 33777937 PMCID: PMC7991106 DOI: 10.3389/fcell.2021.628317] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/22/2021] [Indexed: 12/26/2022] Open
Abstract
The current treatment for ocular pathological angiogenesis mainly focuses on anti-VEGF signals. This treatment has been confirmed as effective despite the unfavorable side effects and unsatisfactory efficiency. Recently, endothelial cell metabolism, especially glycolysis, has been attracting attention as a potential treatment by an increasing number of researchers. Emerging evidence has shown that regulation of endothelial glycolysis can influence vessel sprouting. This new evidence has raised the potential for novel treatment targets that have been overlooked for a long time. In this review, we discuss the process of endothelial glycolysis as a promising target and consider regulation of the enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase as treatment for ocular pathological angiogenesis.
Collapse
Affiliation(s)
- Zi-Yi Zhou
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi’an, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yu-Sheng Wang
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi’an, China
| | - Guo-Rui Dou
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
33
|
Guo Y, Li W, Qian M, Jiang T, Guo P, Du Q, Lin N, Xie X, Wu Z, Lin D, Liu D. D-4F Ameliorates Contrast Media-Induced Oxidative Injuries in Endothelial Cells via the AMPK/PKC Pathway. Front Pharmacol 2021; 11:556074. [PMID: 33658920 PMCID: PMC7917283 DOI: 10.3389/fphar.2020.556074] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/30/2020] [Indexed: 01/23/2023] Open
Abstract
Endothelial dysfunction is involved in the pathophysiological processes of contrast media (CM)–induced acute kidney injury (CI-AKI) after vascular angiography or intervention. Previous study found that apolipoprotein A-I (apoA-I) mimetic peptide, D-4F, alleviates endothelial impairments via upregulating heme oxygenase-1 (HO-1) expression and scavenging excessively generated reactive oxygen species (ROS). However, whether D-4F could ameliorate oxidative injuries in endothelial cells through suppressing ROS production remains unclear. In this study, a representative nonionic iodinated CM, iodixanol, was chosen for the in vitro and in vivo studies. Endothelial cell viability was assayed using micrographs, lactate dehydrogenase (LDH) activity, and cell counting kit-8 (CCK-8). Apoptosis was detected using flow cytometry analysis and caspase-3 activation. Endothelial inflammation was tested using monocyte adhesion assay and adhesion molecule expression. ROS production was detected by measuring the formation of lipid peroxidation malondialdehyde (MDA) through the thiobarbituric acid reactive substance (TBARS) assay. Peroxynitrite (ONOO⁻) formation was tested using the 3-nitrotyrosine ELISA kit. Iodixanol impaired cell viability, promoted vascular cell adhesion molecule-1 (VCAM-1) and intercellular cell adhesion molecule-1 (ICAM-1) expression, and induced cell apoptosis in human umbilical vein endothelial cells (HUVECs). However, D-4F mitigated these injuries. Furthermore, iodixanol induced the phosphorylation of protein kinase C (PKC) beta II, p47, Rac1, and endothelial nitric oxide synthase (eNOS) at Thr495, which elicited ROS release and ONOO⁻ generation. D-4F inhibited NADPH oxidase (NOX) activation, ROS production, and ONOO⁻ formation via the AMP-activated protein kinase (AMPK)/PKC pathway. Additionally, after an intravascular injection of iodixanol in Sprague Dawley rats, iodixanol induced a remarkable inflammatory response in arterial endothelial cells, although significant apoptosis and morphological changes were not observed. D-4F alleviated the vessel inflammation resulting from iodixanol in vivo. Collectively, besides scavenging ROS, D-4F could also suppress ROS production and ONOO⁻ formation through the AMPK/PKC pathway, which ameliorated oxidative injuries in endothelial cells. Hence, D-4F might serve as a potential agent in preventing CI-AKI.
Collapse
Affiliation(s)
- Yansong Guo
- Department of Cardiology, Fujian Provincial Hospital, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Cardiovascular Institute, Fujian Provincial Center for Geriatrics, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Wei Li
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Mingming Qian
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Ting Jiang
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, High-field NMR Research Center, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Ping Guo
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Qian Du
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Na Lin
- Department of Cardiology, Fujian Provincial Hospital, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Cardiovascular Institute, Fujian Provincial Center for Geriatrics, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Xianwei Xie
- Department of Cardiology, Fujian Provincial Hospital, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Cardiovascular Institute, Fujian Provincial Center for Geriatrics, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Zhiyong Wu
- Department of Cardiology, Fujian Provincial Hospital, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Cardiovascular Institute, Fujian Provincial Center for Geriatrics, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Donghai Lin
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, High-field NMR Research Center, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Donghui Liu
- Department of Cardiology, Fujian Provincial Hospital, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Cardiovascular Institute, Fujian Provincial Center for Geriatrics, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China.,Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| |
Collapse
|
34
|
Alhayaza R, Haque E, Karbasiafshar C, Sellke FW, Abid MR. The Relationship Between Reactive Oxygen Species and Endothelial Cell Metabolism. Front Chem 2020; 8:592688. [PMID: 33330380 PMCID: PMC7732658 DOI: 10.3389/fchem.2020.592688] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD) has been the leading cause of death for many decades, highlighting the importance of new research and treatments in the field. The role of hypoxia and subsequent free radical production [reactive oxygen species (ROS)] have become an area of particular interest in CVD. Interestingly, our laboratory and other laboratories have recently reported positive roles of subcellular ROS in modulating endothelial cell (EC) metabolism, proliferation, and angiogenesis. This bidirectional relationship between ROS and EC metabolism, as well as functional changes, continues to be an area of active research. Interestingly, ECs have been shown to rely on anaerobic processes for ATP generation, despite their direct access to oxygen. This paradox has proven to be beneficial as the major reliance on glycolysis produces ATP faster, preserves oxygen, and results in reduced ROS levels in contrast to oxidative phosphorylation. This review will address the relationship between ROS and carbohydrate, lipid, and nitrogen metabolism in ECs, and their effects on EC phenotype such as sprouting angiogenesis.
Collapse
Affiliation(s)
- Raid Alhayaza
- Alfaisal University School of Medicine, Riyadh, Saudi Arabia
| | - Emaan Haque
- Alfaisal University School of Medicine, Riyadh, Saudi Arabia
| | - Catherine Karbasiafshar
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Alpert Medical School, Providence, RI, United States
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Alpert Medical School, Providence, RI, United States
| | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Alpert Medical School, Providence, RI, United States
| |
Collapse
|
35
|
Pericyte-Endothelial Interactions in the Retinal Microvasculature. Int J Mol Sci 2020; 21:ijms21197413. [PMID: 33049983 PMCID: PMC7582747 DOI: 10.3390/ijms21197413] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/25/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
Retinal microvasculature is crucial for the visual function of the neural retina. Pericytes and endothelial cells (ECs) are the two main cellular constituents in the retinal microvessels. Formation, maturation, and stabilization of the micro-vasculatures require pericyte-endothelial interactions, which are perturbed in many retinal vascular disorders, such as retinopathy of prematurity, retinal vein occlusion, and diabetic retinopathy. Understanding the cellular and molecular mechanisms of pericyte-endothelial interaction and perturbation can facilitate the design of therapeutic intervention for the prevention and treatment of retinal vascular disorders. Pericyte-endothelial interactions are indispensable for the integrity and functionality of retinal neurovascular unit (NVU), including vascular cells, retinal neurons, and glial cells. The essential autocrine and paracrine signaling pathways, such as Vascular endothelial growth factor (VEGF), Platelet-derived growth factor subunit B (PDGFB), Notch, Angipointein, Norrin, and Transforming growth factor-beta (TGF-β), have been well characterized for the regulation of pericyte-endothelial interactions in the neo-vessel formation processes (vasculogenesis and angiogenesis) during embryonic development. They also play a vital role in stabilizing and remodeling mature vasculature under pathological conditions. Awry signals, aberrant metabolisms, and pathological conditions, such as oxidative stress and inflammation, can disrupt the communication between pericytes and endothelial cells, thereby resulting in the breakdown of the blood-retinal barrier (BRB) and other microangiopathies. The emerging evidence supports extracellular exosomes' roles in the (mis)communications between the two cell types. This review summarizes the essential knowledge and updates about new advancements in pericyte-EC interaction and communication, emphasizing the retinal microvasculature.
Collapse
|
36
|
Bowen TS, Egginton S. Environmental stress influences mitochondrial metabolism in vascular cells: consequences for angiogenesis. VASCULAR BIOLOGY 2020; 1:H111-H116. [PMID: 32923962 PMCID: PMC7439850 DOI: 10.1530/vb-19-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/01/2019] [Indexed: 11/08/2022]
Abstract
While the important and varied roles that vascular cells play in both health and disease is well recognised, the focus on potential therapeutic targets continually shifts as new players emerge. Here, we outline how mitochondria may be viewed as more than simply energy-generating organelles, but instead as important sentinels of metabolic health and effectors of appropriate responses to physiological challenges.
Collapse
Affiliation(s)
- T Scott Bowen
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Stuart Egginton
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
37
|
Eakin AJ, Mc Erlain T, Burke A, Eaton A, Tipping N, Allocca G, Branco CM. Circulating Levels of Epirubicin Cause Endothelial Senescence While Compromising Metabolic Activity and Vascular Function. Front Cell Dev Biol 2020; 8:799. [PMID: 32974345 PMCID: PMC7466755 DOI: 10.3389/fcell.2020.00799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/28/2020] [Indexed: 12/21/2022] Open
Abstract
Anthracycline-based chemotherapy is a common treatment for cancer patients. Because it is delivered intravenously, endothelial cells are exposed first and to the highest concentrations, prior to diffusion to target cells. Not surprisingly, vascular dysfunction is a consequence of anthracycline therapy. While chemotherapy-induced endothelial damage at administration sites has been investigated, the effects of lower doses encountered by distant microvascular networks has not. The aim of this study was to investigate the impact of epirubicin, a widely used anthracycline, on healthy endothelial cells to elucidate its effects on microvascular physiology. Here, endothelial cells were briefly exposed to low doses of epirubicin to recapitulate levels in circulation following dilution in the blood and compound half-life in circulation. Both immediate and prolonged responses to treatment were assessed to determine changes in endothelial function. Epirubicin caused a decrease in proliferation and viability in hUVEC, with lower doses resulting in a senescent phenotype in a large proportion of cells, accompanied by a significant increase in pro-inflammatory cytokines and a significant decrease in metabolic activity. Epirubicin exposure also impaired endothelial function with delayed wound closure, reduced angiogenic potential and increased monolayer permeability downstream of VE-cadherin internalization. Primary lung endothelial cells obtained from epirubicin-treated mice similarly demonstrated reduced viability and functional impairment. In vivo, epirubicin treatment resulted in persistent reduction in lung vascular density and significantly increased infiltration of myeloid cells. Modulation of endothelial status and inflammatory tissue microenvironment observed in response to low doses of epirubicin may predict risk for long-term secondary pathologies associated with chemotherapy.
Collapse
Affiliation(s)
- Amanda J Eakin
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Tamara Mc Erlain
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Aileen Burke
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Amy Eaton
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Nuala Tipping
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Gloria Allocca
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Cristina M Branco
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
38
|
Motawe ZY, Farsaei F, Abdelmaboud SS, Cuevas J, Breslin JW. Sigma-1 receptor activation-induced glycolytic ATP production and endothelial barrier enhancement. Microcirculation 2020; 27:e12620. [PMID: 32279379 PMCID: PMC7821090 DOI: 10.1111/micc.12620] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/14/2020] [Accepted: 04/02/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE We tested the hypothesis that σ1 modulates endothelial barrier function due to its influence on endothelial bioenergetics. METHODS Cultured HUVEC monolayers were used to model the endothelial barrier. ECIS, Transwell assays, and immunofluorescence labeling of junctional proteins were used to evaluate endothelial barrier function. Endothelial cell bioenergetics was determined using extracellular flux analysis and direct ATP level measurements. The endothelial-specific contribution of σ1 was tested using the σ1-selective agonist, PRE-084, and with targeted knockdown of σ1 expression using siRNA. RESULTS Activation of σ1 with PRE-084 significantly enhanced endothelial barrier function and decreased permeability to albumin and dextran. Knockdown of σ1 with siRNA reduced barrier function and abolished PRE-084-induced endothelial barrier enhancement. PRE-084 upregulated endothelial glycolysis and glycolytic ATP production, but this response was abolished by siRNA-mediated knockdown of σ1 expression. PRE-084 also reduced the degree of endothelial barrier dysfunction caused by the mitochondrial oxidative phosphorylation uncoupler CCCP. CONCLUSION Activation of σ1 enhances endothelial barrier function and modulates the ratio of glycolytic versus mitochondrial ATP production. These novel findings suggest that endothelial σ1 may prove beneficial as a novel therapeutic target for reducing microvascular hyperpermeability and counteracting mitochondrial dysfunction.
Collapse
Affiliation(s)
- Zeinab Y Motawe
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Forouzandeh Farsaei
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Salma S Abdelmaboud
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Javier Cuevas
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jerome W Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
39
|
The Impact of Dietary Supplementation of Whole Foods and Polyphenols on Atherosclerosis. Nutrients 2020; 12:nu12072069. [PMID: 32664664 PMCID: PMC7400924 DOI: 10.3390/nu12072069] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/01/2020] [Accepted: 07/09/2020] [Indexed: 12/16/2022] Open
Abstract
The purpose of this review is to highlight current research on the benefits of supplementation with foods with a diverse polyphenol composition, including fruits, vegetables, nuts, grains, oils, spices, and teas in blunting atherosclerosis. We searched PubMed for publications utilizing whole food or polyphenols prepared from whole foods in Apolipoprotein E (ApoE) or Low-Density Lipoprotein Receptor (LDLR) knockout mice, and identified 73 studies in which plaque was measured. The majority of the studies reported a reduction in plaque. Nine interventions showed no effect, while three using Agaricus blazei mushroom, HYJA-ri-4 rice variety, and safrole-2', 3'-oxide (SFO) increased plaque. The mechanisms by which atherosclerosis was reduced include improved lipid profile, antioxidant status, and cholesterol clearance, and reduced inflammation. Importantly, not all dietary interventions that reduce plaque showed an improvement in lipid profile. Additionally, we found that, out of 73 studies, only 9 used female mice and only 6 compared both sexes. Only one study compared the two models (LDLR vs. ApoE), showing that the treatment worked in one but not the other. Not all supplementations work in both male and female animals, suggesting that increasing the variety of foods with different polyphenol compositions may be more effective in mitigating atherosclerosis.
Collapse
|
40
|
Talepoor AG, Fouladseresht H, Khosropanah S, Doroudchi M. Immune-Inflammation in Atherosclerosis: A New Twist in an Old Tale. Endocr Metab Immune Disord Drug Targets 2020; 20:525-545. [DOI: 10.2174/1871530319666191016095725] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/26/2019] [Accepted: 09/23/2019] [Indexed: 12/27/2022]
Abstract
Background and Objective:Atherosclerosis, a chronic and progressive inflammatory disease, is triggered by the activation of endothelial cells followed by infiltration of innate and adaptive immune cells including monocytes and T cells in arterial walls. Major populations of T cells found in human atherosclerotic lesions are antigen-specific activated CD4+ effectors and/or memory T cells from Th1, Th17, Th2 and Treg subsets. In this review, we will discuss the significance of T cell orchestrated immune inflammation in the development and progression of atherosclerosis.Discussion:Pathogen/oxidative stress/lipid induced primary endothelial wound cannot develop to a full-blown atherosclerotic lesion in the absence of chronically induced inflammation. While the primary inflammatory response might be viewed as a lone innate response, the persistence of such a profound response over time must be (and is) associated with diverse local and systemic T cell responses. The interplay between T cells and innate cells contributes to a phenomenon called immuneinflammation and has an impact on the progression and outcome of the lesion. In recent years immuneinflammation, an old term, has had a comeback in connecting the puzzle pieces of chronic inflammatory diseases.Conclusion:Taking one-step back and looking from afar at the players of immune-inflammation may help us provide a broader perspective of these complicated interactions. This may lead to the identification of new drug targets and the development of new therapies as well as preventative measures.
Collapse
Affiliation(s)
- Atefe Ghamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Fouladseresht
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahdad Khosropanah
- Department of Cardiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
41
|
Shimizu Y, Nakamura Y, Horibata Y, Fujimaki M, Hayashi K, Uchida N, Morita H, Arai R, Chibana K, Takemasa A, Sugimoto H. Imaging of lysophosphatidylcholine in an induced pluripotent stem cell-derived endothelial cell network. Regen Ther 2020; 14:299-305. [PMID: 32462058 PMCID: PMC7240204 DOI: 10.1016/j.reth.2020.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/01/2020] [Accepted: 03/11/2020] [Indexed: 12/31/2022] Open
Abstract
Introduction Vascular endothelial cell disorders are closely related to cardiovascular disease (CVD) and pulmonary diseases. Abnormal lipid metabolism in the endothelium leads to changes in cell signalling, and the expression of genes related to immunity and inflammation. It is therefore important to investigate the pathophysiology of vascular endothelial disorders in terms of lipid metabolism, using a disease model of endothelium. Methods Human induced pluripotent stem cell-derived endothelial cells (iECs) were cultured on a matrigel to form an iEC network. Lipids in the iEC network were investigated by matrix-assisted laser desorption/ionization (MALDI) time-of-flight (TOF) imaging mass spectrometry (IMS) analysis. Ion fragments obtained by mass spectrometry were analysed using an infusion method, involving precursor ion scanning with fragment ion. Results The MALDI TOF IMS analysis revealed co-localized intensity of peaks at m/z 592.1 and 593.1 in the iEC network. Tandem mass spectrometry (MS/MS) analysis by MALDI-imaging, in conjunction with precursor ion scanning using an infusion method with lipid extracts, identified that these precursor ions were lysophosphatidylcholine (LPC) (22:5) and its isotype. Conclusion The MALDI-imaging analysis showed that LPC (22:5) was abundant in an iEC network. As an in vitro test model for disease and potential therapy, present analysis methods using MALDI-imaging combined with, for example, mesenchymal stem cells (MSC) to a disease derived iEC network may be useful in revealing the changes in the amount and distribution of lipids under various stimuli.
Collapse
Affiliation(s)
- Yasuo Shimizu
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Yusuke Nakamura
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Yasuhiro Horibata
- Department of Biochemistry, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Mio Fujimaki
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Keitaro Hayashi
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Nobuhiko Uchida
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Hiroko Morita
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Ryo Arai
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Kazuyuki Chibana
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Akihiro Takemasa
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Hiroyuki Sugimoto
- Department of Biochemistry, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| |
Collapse
|
42
|
Green HLH, Brewer AC. Dysregulation of 2-oxoglutarate-dependent dioxygenases by hyperglycaemia: does this link diabetes and vascular disease? Clin Epigenetics 2020; 12:59. [PMID: 32345373 PMCID: PMC7189706 DOI: 10.1186/s13148-020-00848-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023] Open
Abstract
The clinical, social and economic burden of cardiovascular disease (CVD) associated with diabetes underscores an urgency for understanding the disease aetiology. Evidence suggests that the hyperglycaemia associated with diabetes is, of itself, causal in the development of endothelial dysfunction (ED) which is recognised to be the critical determinant in the development of CVD. It is further recognised that epigenetic modifications associated with changes in gene expression are causal in both the initiation of ED and the progression to CVD. Understanding whether and how hyperglycaemia induces epigenetic modifications therefore seems crucial in the development of preventative treatments. A mechanistic link between energy metabolism and epigenetic regulation is increasingly becoming explored as key energy metabolites typically serve as substrates or co-factors for epigenetic modifying enzymes. Intriguing examples are the ten-eleven translocation and Jumonji C proteins which facilitate the demethylation of DNA and histones respectively. These are members of the 2-oxoglutarate-dependent dioxygenase superfamily which require the tricarboxylic acid metabolite, α-ketoglutarate and molecular oxygen (O2) as substrates and Fe (II) as a co-factor. An understanding of precisely how the biochemical effects of high glucose exposure impact upon cellular metabolism, O2 availability and cellular redox in endothelial cells (ECs) may therefore elucidate (in part) the mechanistic link between hyperglycaemia and epigenetic modifications causal in ED and CVD. It would also provide significant proof of concept that dysregulation of the epigenetic landscape may be causal rather than consequential in the development of pathology.
Collapse
Affiliation(s)
- Hannah L H Green
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| | - Alison C Brewer
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK.
| |
Collapse
|
43
|
Abstract
Coronary heart disease (CHD) is the most common and serious illness in the world and has been researched for many years. However, there are still no real effective ways to prevent and save patients with this disease. When patients present with myocardial infarction, the most important step is to recover ischemic prefusion, which usually is accomplished by coronary artery bypass surgery, coronary artery intervention (PCI), or coronary artery bypass grafting (CABG). These are invasive procedures, and patients with extensive lesions cannot tolerate surgery. It is, therefore, extremely urgent to search for a noninvasive way to save ischemic myocardium. After suffering from ischemia, cardiac or skeletal muscle can partly recover blood flow through angiogenesis (de novo capillary) induced by hypoxia, arteriogenesis, or collateral growth (opening and remodeling of arterioles) triggered by dramatical increase of fluid shear stress (FSS). Evidence has shown that both of them are regulated by various crossed pathways, such as hypoxia-related pathways, cellular metabolism remodeling, inflammatory cells invasion and infiltration, or hemodynamical changes within the vascular wall, but still they do not find effective target for regulating revascularization at present. 5′-Adenosine monophosphate-activated protein kinase (AMPK), as a kinase, is not only an energy modulator but also a sensor of cellular oxygen-reduction substances, and many researches have suggested that AMPK plays an essential role in revascularization but the mechanism is not completely understood. Usually, AMPK can be activated by ADP or AMP, upstream kinases or other cytokines, and pharmacological agents, and then it phosphorylates key molecules that are involved in energy metabolism, autophagy, anti-inflammation, oxidative stress, and aging process to keep cellular homeostasis and finally keeps cell normal activity and function. This review makes a summary on the subunits, activation and downstream targets of AMPK, the mechanism of revascularization, the effects of AMPK in endothelial cells, angiogenesis, and arteriogenesis along with some prospects.
Collapse
|
44
|
Bartman CM, Eckle T. Circadian-Hypoxia Link and its Potential for Treatment of Cardiovascular Disease. Curr Pharm Des 2020; 25:1075-1090. [PMID: 31096895 DOI: 10.2174/1381612825666190516081612] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/03/2019] [Indexed: 12/29/2022]
Abstract
Throughout the evolutionary time, all organisms and species on Earth evolved with an adaptation to consistent oscillations of sunlight and darkness, now recognized as 'circadian rhythm.' Single-cellular to multisystem organisms use circadian biology to synchronize to the external environment and provide predictive adaptation to changes in cellular homeostasis. Dysregulation of circadian biology has been implicated in numerous prevalent human diseases, and subsequently targeting the circadian machinery may provide innovative preventative or treatment strategies. Discovery of 'peripheral circadian clocks' unleashed widespread investigations into the potential roles of clock biology in cellular, tissue, and organ function in healthy and diseased states. Particularly, oxygen-sensing pathways (e.g. hypoxia inducible factor, HIF1), are critical for adaptation to changes in oxygen availability in diseases such as myocardial ischemia. Recent investigations have identified a connection between the circadian rhythm protein Period 2 (PER2) and HIF1A that may elucidate an evolutionarily conserved cellular network that can be targeted to manipulate metabolic function in stressed conditions like hypoxia or ischemia. Understanding the link between circadian and hypoxia pathways may provide insights and subsequent innovative therapeutic strategies for patients with myocardial ischemia. This review addresses our current understanding of the connection between light-sensing pathways (PER2), and oxygen-sensing pathways (HIF1A), in the context of myocardial ischemia and lays the groundwork for future studies to take advantage of these two evolutionarily conserved pathways in the treatment of myocardial ischemia.
Collapse
Affiliation(s)
- Colleen Marie Bartman
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, Graduate Training Program in Cell Biology, Stem Cells, and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Tobias Eckle
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, Graduate Training Program in Cell Biology, Stem Cells, and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
45
|
Bou Khzam L, Son NH, Mullick AE, Abumrad NA, Goldberg IJ. Endothelial cell CD36 deficiency prevents normal angiogenesis and vascular repair. Am J Transl Res 2020; 12:7737-7761. [PMID: 33437358 PMCID: PMC7791529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/23/2020] [Indexed: 03/16/2023]
Abstract
Endothelial cells (ECs) maintain vascular integrity and mediate vascular repair and angiogenesis, by which new blood vessels are formed from pre-existing blood vessels. Hyperglycemia has been shown to increase EC angiogenic potential. However, few studies have investigated effects of fatty acids (FAs) on EC angiogenesis. Cluster of differentiation 36 (CD36) is a FA transporter expressed by ECs, but its role in EC proliferation, migration, and angiogenesis is unknown. We sought to determine if circulating FAs regulate angiogenic function in a CD36-dependent manner. CD36-dependent effects of FAs on EC proliferation and migration of mouse heart ECs (MHECs) and lung ECs (MLECs) were studied. We used both silencing RNA and antisense oligonucleotides to reduce CD36 expression. Oleic acid (OA) did not affect EC proliferation, but significantly increased migration of ECs in wound healing experiments. CD36 knockdown prevented OA-induced increases in wound healing potential. In EC transwell migration experiments, OA increased recruitment and migration of ECs, an effect abolished by CD36 knockdown. Phospho-AMP-activated protein kinase (AMPK) increased in MHECs exposed to OA in a CD36-dependent manner. To test whether in vivo CD36 affects angiogenesis, we studied 21-day recovery in post-hindlimb ischemia. EC-specific CD36 knockout mice had reduced blood flow recovery as assessed by laser Doppler imaging. EC content in post-ischemic muscle, assessed from CD31 expression, increased in ischemic muscle of control mice. However, mice with EC-specific CD36 deletion lacked the increase in CD31 and matrix metalloprotease 9 expression observed in controls. EC expression of CD36 and its function in FA uptake modulate angiogenic function and response to ischemia, likely due to reduced activation of the AMPK pathway.
Collapse
Affiliation(s)
- Lara Bou Khzam
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, NYU Langone Health New York, NY, USA
| | - Ni-Huiping Son
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, NYU Langone Health New York, NY, USA
| | | | - Nada A Abumrad
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Ira J Goldberg
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, NYU Langone Health New York, NY, USA
| |
Collapse
|
46
|
Dikalova AE, Pandey A, Xiao L, Arslanbaeva L, Sidorova T, Lopez MG, Billings FT, Verdin E, Auwerx J, Harrison DG, Dikalov SI. Mitochondrial Deacetylase Sirt3 Reduces Vascular Dysfunction and Hypertension While Sirt3 Depletion in Essential Hypertension Is Linked to Vascular Inflammation and Oxidative Stress. Circ Res 2019; 126:439-452. [PMID: 31852393 DOI: 10.1161/circresaha.119.315767] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
RATIONALE Hypertension represents a major risk factor for stroke, myocardial infarction, and heart failure and affects 30% of the adult population. Mitochondrial dysfunction contributes to hypertension, but specific mechanisms are unclear. The mitochondrial deacetylase Sirt3 (Sirtuin 3) is critical in the regulation of metabolic and antioxidant functions which are associated with hypertension, and cardiovascular disease risk factors diminish Sirt3 level. OBJECTIVE We hypothesized that reduced Sirt3 expression contributes to vascular dysfunction in hypertension, but increased Sirt3 protects vascular function and decreases hypertension. METHODS AND RESULTS To test the therapeutic potential of targeting Sirt3 expression, we developed new transgenic mice with global Sirt3OX (Sirt3 overexpression), which protects from endothelial dysfunction, vascular oxidative stress, and hypertrophy and attenuates Ang II (angiotensin II) and deoxycorticosterone acetate-salt induced hypertension. Global Sirt3 depletion in Sirt3-/- mice results in oxidative stress due to hyperacetylation of mitochondrial superoxide dismutase (SOD2), increases HIF1α (hypoxia-inducible factor-1), reduces endothelial cadherin, stimulates vascular hypertrophy, increases vascular permeability and vascular inflammation (p65, caspase 1, VCAM [vascular cell adhesion molecule-1], ICAM [intercellular adhesion molecule-1], and MCP1 [monocyte chemoattractant protein 1]), increases inflammatory cell infiltration in the kidney, reduces telomerase expression, and accelerates vascular senescence and age-dependent hypertension; conversely, increased Sirt3 expression in Sirt3OX mice prevents these deleterious effects. The clinical relevance of Sirt3 depletion was confirmed in arterioles from human mediastinal fat in patients with essential hypertension showing a 40% decrease in vascular Sirt3, coupled with Sirt3-dependent 3-fold increases in SOD2 acetylation, NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) activity, VCAM, ICAM, and MCP1 levels in hypertensive subjects compared with normotensive subjects. CONCLUSIONS We suggest that Sirt3 depletion in hypertension promotes endothelial dysfunction, vascular hypertrophy, vascular inflammation, and end-organ damage. Our data support a therapeutic potential of targeting Sirt3 expression in vascular dysfunction and hypertension.
Collapse
Affiliation(s)
- Anna E Dikalova
- From the Vanderbilt University Medical Center, Nashville, TN (A.E.D., A.P., L.X., L.A., T.S., M.G.L., F.T.B., D.G.H., S.I.D.)
| | - Arvind Pandey
- From the Vanderbilt University Medical Center, Nashville, TN (A.E.D., A.P., L.X., L.A., T.S., M.G.L., F.T.B., D.G.H., S.I.D.)
| | - Liang Xiao
- From the Vanderbilt University Medical Center, Nashville, TN (A.E.D., A.P., L.X., L.A., T.S., M.G.L., F.T.B., D.G.H., S.I.D.)
| | - Liaisan Arslanbaeva
- From the Vanderbilt University Medical Center, Nashville, TN (A.E.D., A.P., L.X., L.A., T.S., M.G.L., F.T.B., D.G.H., S.I.D.)
| | - Tatiana Sidorova
- From the Vanderbilt University Medical Center, Nashville, TN (A.E.D., A.P., L.X., L.A., T.S., M.G.L., F.T.B., D.G.H., S.I.D.)
| | - Marcos G Lopez
- From the Vanderbilt University Medical Center, Nashville, TN (A.E.D., A.P., L.X., L.A., T.S., M.G.L., F.T.B., D.G.H., S.I.D.)
| | - Frederic T Billings
- From the Vanderbilt University Medical Center, Nashville, TN (A.E.D., A.P., L.X., L.A., T.S., M.G.L., F.T.B., D.G.H., S.I.D.)
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA (E.V.)
| | - Johan Auwerx
- Ecole Polytechnique Fédérale de Lausanne, Switzerland (J.A.)
| | - David G Harrison
- From the Vanderbilt University Medical Center, Nashville, TN (A.E.D., A.P., L.X., L.A., T.S., M.G.L., F.T.B., D.G.H., S.I.D.)
| | - Sergey I Dikalov
- From the Vanderbilt University Medical Center, Nashville, TN (A.E.D., A.P., L.X., L.A., T.S., M.G.L., F.T.B., D.G.H., S.I.D.)
| |
Collapse
|
47
|
Tiemeier GL, Wang G, Dumas SJ, Sol WMPJ, Avramut MC, Karakach T, Orlova VV, van den Berg CW, Mummery CL, Carmeliet P, van den Berg BM, Rabelink TJ. Closing the Mitochondrial Permeability Transition Pore in hiPSC-Derived Endothelial Cells Induces Glycocalyx Formation and Functional Maturation. Stem Cell Reports 2019; 13:803-816. [PMID: 31680061 PMCID: PMC6895683 DOI: 10.1016/j.stemcr.2019.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 10/04/2019] [Accepted: 10/05/2019] [Indexed: 11/23/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are used to study organogenesis and model disease as well as being developed for regenerative medicine. Endothelial cells are among the many cell types differentiated from hiPSCs, but their maturation and stabilization fall short of that in adult endothelium. We examined whether shear stress alone or in combination with pericyte co-culture would induce flow alignment and maturation of hiPSC-derived endothelial cells (hiPSC-ECs) but found no effects comparable with those in primary microvascular ECs. In addition, hiPSC-ECs lacked a luminal glycocalyx, critical for vasculature homeostasis, shear stress sensing, and signaling. We noted, however, that hiPSC-ECs have dysfunctional mitochondrial permeability transition pores, resulting in reduced mitochondrial function and increased reactive oxygen species. Closure of these pores by cyclosporine A improved EC mitochondrial function but also restored the glycocalyx such that alignment to flow took place. These results indicated that mitochondrial maturation is required for proper hiPSC-EC functionality. hiPSC-ECs lack a functional glycocalyx and fail to align to flow hiPSC-ECs have reduced mitochondrial function and increased leakage of ROS Closing the mPTP with cyclosporine A induces mitochondrial maturation Improved mitochondrial function restores the glycocalyx and alignment to flow
Collapse
Affiliation(s)
- Gesa L Tiemeier
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gangqi Wang
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sébastien J Dumas
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Wendy M P J Sol
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - M Cristina Avramut
- Department of Cell and Chemical Biology, Section Electron Microscopy, Leiden University Medical Center, Leiden, The Netherlands
| | - Tobias Karakach
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cathelijne W van den Berg
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Bernard M van den Berg
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ton J Rabelink
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
48
|
Huang H, Lennikov A, Saddala MS, Gozal D, Grab DJ, Khalyfa A, Fan L. Placental growth factor negatively regulates retinal endothelial cell barrier function through suppression of glucose-6-phosphate dehydrogenase and antioxidant defense systems. FASEB J 2019; 33:13695-13709. [PMID: 31585507 DOI: 10.1096/fj.201901353r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We report that placental growth factor (PlGF) negatively affects the endothelial cell (EC) barrier function through a novel regulatory mechanism. The PlGF mAb promotes (but recombinant protein disrupts) EC barrier function, thus affecting the barrier-forming protein levels, membrane distribution, and EC monolayer impedance by the electrical cell-impedance sensing system, Western blot, and immunofluorescence staining. RNA sequencing-based transcriptome analysis identified the up-regulation of the pentose phosphate pathway (PPP) and the antioxidant defense protein by PlGF blockade. The PlGF and PlGF/VEGF dimers (but not VEGF-A) down-regulated the protein expression of glucose-6-phosphate dehydrogenase (G6PD) and peroxiredoxin (PRDX). G6PD inhibition and gene silencing (small interfering RNA) abolished the beneficial effects of PlGF inhibition on EC barrier function and PRDX3/6 protein expression. VEGF receptor (VEGFR)1 or VEGFR2 blockade prevented the inhibitory effect of PlGF on G6PD protein expression and EC barrier function. The PRDX6 played dual roles in EC barrier function through glutathione peroxidase and phospholipase A2 activity. In sum, PlGF negatively regulates EC barrier function through the activation of VEGFR1 and VEGFR2 and the suppression of the G6PD/PPP and the antioxidant pathways.-Huang, H., Lennikov, A., Saddala, M. S., Gozal, D., Grab, D. J., Khalyfa, A., Fan, L. Placental growth factor negatively regulates endothelial cell barrier function through suppression of glucose-6-phosphate dehydrogenase and antioxidant defense systems.
Collapse
Affiliation(s)
- Hu Huang
- Department of Ophthalmology, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Anton Lennikov
- Department of Ophthalmology, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Madhu Sudhana Saddala
- Department of Ophthalmology, University of Missouri-Columbia, Columbia, Missouri, USA
| | - David Gozal
- Child Health Research Institute, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Dennis J Grab
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA.,Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Abdelnaby Khalyfa
- Child Health Research Institute, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Lijuan Fan
- Department of Ophthalmology, University of Missouri-Columbia, Columbia, Missouri, USA
| |
Collapse
|
49
|
von Meyenn L, Bertschi NL, Schlapbach C. Targeting T Cell Metabolism in Inflammatory Skin Disease. Front Immunol 2019; 10:2285. [PMID: 31608068 PMCID: PMC6769046 DOI: 10.3389/fimmu.2019.02285] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/10/2019] [Indexed: 12/31/2022] Open
Abstract
A properly functioning T cell compartment is crucial to protect the host from infections, tumors, and environmental substances. In recent years, it has become increasingly clear that the processes underlying proper T cell activation, proliferation, and differentiation require well-tuned and dynamic changes in T cell metabolism. Thus, proper metabolic reprogramming in T cells is crucial to ensure proper immunity in the context of infection and anti-tumor immunity. Conversely, aberrant regulation of T cell metabolism can impair T cell function and thereby contribute to T cell-mediated disease. In this review, the relevance of recent insights into T cell metabolism for prototypical T cell-mediated skin diseases will be discussed and their therapeutic potential will be outlined. First, the major modules of T cell metabolism are summarized. Then, the importance of T cell metabolism for T cell-mediated skin diseases such as psoriasis and allergic contact dermatitis is discussed, based on the current state of our understanding thereof. Finally, novel therapeutic opportunities for inflammatory skin disease that might emerge from investigations in T cell metabolism are outlined.
Collapse
Affiliation(s)
| | | | - Christoph Schlapbach
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
50
|
(Energetic metabolism of endothelial cell). COR ET VASA 2019. [DOI: 10.33678/cor.2019.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|