1
|
Roldán Gallardo FF, Martínez Piñerez DE, Reinarz Torrado KF, Berg GA, Herzfeld JD, Da Ros VG, López Seoane M, Maldonado CA, Quintar AA. Extracellular Vesicles Contribute to Oxidized LDL-Induced Stromal Cell Proliferation in Benign Prostatic Hyperplasia. BIOLOGY 2024; 13:827. [PMID: 39452137 PMCID: PMC11504470 DOI: 10.3390/biology13100827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Clinical and experimental evidence has linked Benign Prostatic Hyperplasia (BPH) with dyslipidemic and hypercholesterolemic conditions, though the underlying cellular mechanisms remain unclear. This study investigates the impact of dyslipidemia, specifically oxidized LDL (OxLDL), on prostatic stromal cell proliferation and the release of extracellular vesicles (EVs). METHODS Mice were fed a high-fat diet, and human prostatic stromal cells (HPSCs) were treated with OxLDL. Proliferation assays and EV characterization were performed to assess the role of EVs in BPH progression. RESULTS Pro-atherogenic conditions significantly increased cell proliferation in both murine prostatic cells and HPSCs. Treatment with metformin effectively inhibited OxLDL-induced proliferation. Additionally, OxLDL stimulated the production and release of pro-proliferative EVs by HPSCs, which further promoted cellular proliferation. CONCLUSIONS The findings suggest that dyslipidemia drives prostatic stromal cell proliferation and EV secretion, contributing to BPH progression. Metformin demonstrates potential as a therapeutic agent to mitigate these effects, offering insight into novel strategies for BPH management. This study highlights the complex interaction between dyslipidemia, cell proliferation, and extracellular communication in the context of BPH pathogenesis.
Collapse
Affiliation(s)
- Franco F. Roldán Gallardo
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (F.F.R.G.)
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, Argentina
| | - Daniel E. Martínez Piñerez
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (F.F.R.G.)
| | - Kevin F. Reinarz Torrado
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (F.F.R.G.)
| | - Gabriela A. Berg
- Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires 1000, Argentina
| | - Jael D. Herzfeld
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires 1000, Argentina
| | - Vanina G. Da Ros
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires 1000, Argentina
| | | | - Cristina A. Maldonado
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (F.F.R.G.)
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, Argentina
| | - Amado A. Quintar
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (F.F.R.G.)
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, Argentina
| |
Collapse
|
2
|
Liu S, Zhang Y, Ma X, Zhan C, Ding N, Shi M, Zhang W, Yang S. Protective effects of engineered Lactobacillus crispatus strains expressing G-CSF on thin endometrium of mice. Hum Reprod 2024; 39:2305-2319. [PMID: 39178354 DOI: 10.1093/humrep/deae190] [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: 11/18/2023] [Revised: 06/26/2024] [Indexed: 08/25/2024] Open
Abstract
STUDY QUESTION Does recombinant Lactobacillus expressing granulocyte colony-stimulating factor (G-CSF) have a better protective effect than the current treatment of thin endometrium (TE)? SUMMARY ANSWER This study suggested that the intrauterine injection of Lactobacillus crispastus (L. crispastus)-pPG612-G-CSF has a positive effect on preventing TE induced by 95% alcohol in mice. WHAT IS KNOWN ALREADY TE has a negative impact on the success rate of ART in patients, and is usually caused by intrauterine surgery, endometrial infection, or hormone drugs. Exogenous G-CSF can promote endometrial vascular remodelling and increase endometrial receptivity and the embryo implantation rate. Moreover, Lactobacillus plays a crucial role in maintaining and regulating the local microecological balance of the reproductive tract, and it could be a delivery carrier of the endometrial repair drug G-CSF. STUDY DESIGN, SIZE, DURATION We constructed engineered L. crispastus strains expressing G-CSF. The mice were divided into five groups: (i) Control group (C, n = 28), uteri were treated with preheated saline solution via intrauterine injection on the third and sixth day of oestrus; (ii) Model group (M, n = 35), where uteri were treated with 95% alcohol on the third day of oestrus and preheated saline solution on the sixth day of oestrus via intrauterine injection; (iii) L. crispatus-pPG612-treatment group (L, n = 45), where uteri were treated with 95% alcohol on the third day of oestrus and 0.1 ml × 108 CFU/ml L. crispatus-pPG612 on the sixth day of oestrus via intrauterine injection; (iv) L. crispatus-pPG612-treatment group (LG, n = 45), where uteri were treated with 95% alcohol on the third day of oestrus and 0.1 ml × 108 CFU/ml L. crispatus-pPG612-G-CSF on the sixth day of oestrus via intrauterine injection; (v) G-CSF-treatment group (G, n = 52), where uteri were treated with 95% alcohol on the third day of oestrus and 30 µg/kg G-CSF on the sixth day of oestrus via intrauterine injection. Then, we compared the effects of L. crispastus, L. crispatus-pPG612-G-CSF and G-CSF on endometrial thickness, angiogenesis, fibrosis, and inflammation in the TE mouse. PARTICIPANTS/MATERIALS, SETTING, METHODS We collected uterine tissues for haematoxylin-eosin staining, immunohistochemical staining, Western blot and RT-PCR, as well as serum for ELISA and uterine flushing solution for high-throughput sequencing. MAIN RESULTS AND THE ROLE OF CHANCE Compared with those in the M group (the mice of the group were intrauterine injected 95% alcohol and treated with saline solution), the L. crispatus-pPG612-G-CSF strain increased the thickness of the endometrium (P < 0.001) and the number of blood vessels and glands (both P < 0.001), enhanced the expression of cytokeratin 19 (CK19) (P < 0.001), vimentin (Vim) (P < 0.001), vascular endothelial growth factor-A (P < 0.001), and CD34 (P < 0.001), and decreased fibrosis levels (P = 0.004). In addition, the high-throughput sequencing results indicated that the L. crispatus-pPG612-G-CSF strain could decrease the abundance of Pseudomonas (P = 0.044) and Actinomyces spp. (P = 0.094) in TE mice and increased the average number of embryos (P = 0.036). Finally, the L. crispatus-pPG612-G-CSF strain was preliminarily confirmed to activate the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) signalling pathway and enhance the mRNA expression of hypoxia-inducible factor-1α (P < 0.001), vascular endometrial growth factor (P = 0.003), and endothelial cell nitric oxide synthase (P = 0.003) in mouse uterine tissue. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Therapy with the L. crispatus-pPG612-G-CSF strain has tremendous potential to accelerate the reparative processes of TE. However, we have reported only the expression of genes and proteins related to the PI3K/AKT pathway, and numerous other mechanisms may also be involved in the restoration of the endometrium by L. crispatus-pPG612-G-CSF. WIDER IMPLICATIONS OF THE FINDINGS The results from the study provide new ideas and suggest new methods for TE treatment. STUDY FUNDING/COMPETING INTEREST(S) This work was financially supported by the Project of Science and Technology Development Plan of Jilin Province (grant number 20210101232JC), the Science and Technology Plan Item of Jilin Provincial Education Department (grant number JT53101022010), and the Doctoral Research Start-up Fund of Jilin Medical University (grant numbers JYBS2021014LK and 2022JYBS006KJ). The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.
Collapse
Affiliation(s)
- Shuang Liu
- Reproductive Immunology Laboratory, Basic Medical College, Jilin Medical University, Jilin, China
- Histology Development Laboratory, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yingnan Zhang
- Chronic Disease Laboratory, School of Public Health, Jilin Medical University, Jilin, China
- Department of Biology, College of Life Science, Changchun Sci-Tech University, Changchun, China
| | - Xin Ma
- Histology Development Laboratory, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Chenglin Zhan
- Histology Development Laboratory, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Ning Ding
- Histology Development Laboratory, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Mai Shi
- Chronic Disease Laboratory, School of Public Health, Jilin Medical University, Jilin, China
| | - Wei Zhang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jilin Medical University, Jilin, China
| | - Shubao Yang
- Reproductive Immunology Laboratory, Basic Medical College, Jilin Medical University, Jilin, China
| |
Collapse
|
3
|
Xian Y, Wang X, Yu Y, Chen X. The mechanism of EGFL7 regulating neovascularization in diabetic retinopathy through the PI3K/AKT/VEGFA pathway. Life Sci 2024; 340:122483. [PMID: 38307238 DOI: 10.1016/j.lfs.2024.122483] [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: 10/11/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
Diabetic retinopathy (DR) is a blinding disease caused by diabetes, characterized by neovascularization of the retina. The aim of this study was to investigate the roles of epidermal growth factor-like structural domain 7 (EGFL7) on human retinal vascular endothelial cells (HRECS) and retinas from rats with DR. An in vitro model of DR was established through culturing HRECS in high glucose. The in vivo model of DR was established by injecting SD rats with streptozotocin (STZ) to induce diabetes. The differences in the expressed levels of EGFL7, PI3K, AKT, P-AKT and VEGFA in high-glucose cultured cells and retinal tissues of diabetic rats were detected in compared to those in the control group. Stable EGFL7 knockdown cell lines were generated by transfecting HRECS with lentiviral vectors and the effects of EGFL7 knockdown on angiogenesis, cell migration and proliferation were investigated. The results showed that EGFL7, PI3K, P-AKT and VEGFA was increased in cells and tissues under high glucose conditions. Knockdown of EGFL7 downregulated the proliferation, migration and angiogenesis capacity of HRECS, and blocked the PI3K/AKT/VEGFA signaling pathway. Furthermore, overexpression of PI3K reversed the effects of EGFL7 inhibition. These findings provide new ideas for the treatment of neovascularisation in DR.
Collapse
Affiliation(s)
- Yang Xian
- Department of Ophthalmology, Shengjing Hospital of China Medical University, China
| | - XingLi Wang
- Department of Ophthalmology, Shengjing Hospital of China Medical University, China
| | - Yong Yu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, China
| | - XiaoLong Chen
- Department of Ophthalmology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
4
|
Yuan Z, Tian Y, Zhang C, Wang M, Xie J, Wang C, Huang J. Integration of systematic review, lipidomics with experiment verification reveals abnormal sphingolipids facilitate diabetic retinopathy by inducing oxidative stress on RMECs. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159382. [PMID: 37659619 DOI: 10.1016/j.bbalip.2023.159382] [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: 05/05/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023]
Abstract
OBJECTIVE This study aims to explore the potential biomarkers in the development of diabetes mellitus (DM) into diabetic retinopathy (DR). METHODS Systematic review of diabetic metabolomics was used to screen the differential metabolites and related pathways during the development of DM. Non-targeted lipidomics of rat plasma was performed to explore the differential metabolites in the development of DM into DR in vivo. To verify the effects of differential metabolites in inducing retinal microvascular endothelial cells (RMECs) injury by increasing oxidative stress, high glucose medium containing differential metabolites was used to induce rat RMECs injury and cell viability, malondialdehyde (MDA) contents, superoxide dismutase (SOD) activities, reactive oxygen species (ROS) levels and mitochondrial membrane potential (MMP) were evaluated in vitro. Network pharmacology was performed to explore the potential mechanism of differential metabolites in inducing DR. RESULTS Through the systematic review, 148 differential metabolites were obtained and the sphingolipid metabolic pathway attracted our attention. Plasma non-targeted lipidomics found that sphingolipids were accompanied by the development of DM into DR. In vitro experiments showed sphinganine and sphingosine-1-phosphate aggravated rat RMECs injury induced by high glucose, further increased MDA and ROS levels, and further decreased SOD activities and MMP. Network pharmacology revealed sphinganine and sphingosine-1-phosphate may induce DR by regulating the AGE-RAGE and HIF-1 signaling pathways. CONCLUSIONS Integrated systematic review, lipidomics and experiment verification reveal that abnormal sphingolipid metabolism facilitates DR by inducing oxidative stress on RMECs. Our study could provide the experimental basis for finding potential biomarkers for the diagnosis and treatment of DR.
Collapse
Affiliation(s)
- Zhenshuang Yuan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yue Tian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Cong Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingshuang Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jiaqi Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Can Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Jianmei Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
5
|
Zou D, Yang P, Liu J, Dai F, Xiao Y, Zhao A, Huang N. Constructing Mal-Efferocytic Macrophage Model and Its Atherosclerotic Spheroids and Rat Model for Therapeutic Evaluation. Adv Biol (Weinh) 2023; 7:e2200277. [PMID: 36721069 DOI: 10.1002/adbi.202200277] [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: 10/11/2022] [Revised: 11/27/2022] [Indexed: 02/02/2023]
Abstract
Efferocytosis, responsible for apoptotic cell clearance, is an essential factor against atherosclerosis. It is reported that efferocytosis is severely impaired in fibroatheroma, especially in vulnerable thin cap fibroatheroma. However, there is a shortage of studies on efferocytosis defects in cell and animal models. Here, the impacts of oxidized low density lipoprotein (ox-LDL) and glut 1 inhibitor (STF31) on efferocytosis of macrophages are studied, and an evaluation system is constructed. Through regulating the cell ratios and stimulus, three types of atherosclerotic spheroids are fabricated, and a necrotic core emerges with surrounding apoptotic cells. Rat models present a similar phenomenon in that substantial apoptotic cells are uncleared in time in vulnerable plaque, and the model period is shortened to 7 weeks. Mechanism studies reveal that ox-LDL, through mRNA and miRNA modulation, downregulates efferocytosis receptor (PPARγ/LXRα/MerTK), internalization molecule (SLC29a1), and upregulates the competitive receptor CD300a that inhibits efferocytosis receptor-ligand binding process. The foam cell differentiation has also confirmed that CD36 and Lp-PLA2 levels are significantly elevated, and macrophages present an interesting transition into prothrombic phenotype. Collectively, the atherosclerotic models featured by efferocytosis defect provide a comprehensive platform to evaluate the efficacy of medicine and biomaterials for atherosclerosis treatment.
Collapse
Affiliation(s)
- Dan Zou
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Ping Yang
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Jianan Liu
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Fanfan Dai
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Yangyang Xiao
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Ansha Zhao
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Nan Huang
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| |
Collapse
|
6
|
Varghese DS, Oommen D, John A, Ali BR. GRP78/BiP alleviates oxLDL-induced hepatotoxicity in familial hypercholesterolemia caused by missense variants of LDLR in a HepG2 cellular model. Lipids Health Dis 2023; 22:69. [PMID: 37248472 DOI: 10.1186/s12944-023-01835-x] [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: 02/22/2023] [Accepted: 05/13/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND AND AIMS The accumulation of misfolded proteins, encoded by genetic variants of functional genes leads to Endoplasmic Reticulum (ER) stress, which is a critical consequence in human disorders such as familial hypercholesterolemia, cardiovascular and hepatic diseases. In addition to the identification of ER stress as a contributing factor to pathogenicity, extensive studies on the role of oxidized Low-Density Lipoprotein (oxLDL) and its ill effects in expediting cardiovascular diseases and other metabolic comorbidities are well documented. However, the current understanding of its role in hepatic insults needs to be revised. This study elucidates the molecular mechanisms underlying the progression of oxLDL and ER stress-induced cytotoxicity in HepG2. METHODS HepG2 cells stably expressing wild-type Low-Density lipoprotein receptor (WT-LDLR) and missense variants of LDLR that are pathogenically associated with familial hypercholesterolemia were used as the in vitro models. The relative mRNA expression and protein profiles of ER stress sensors, inflammatory and apoptotic markers, together with cytotoxic assays and measurement of mitochondrial membrane potential, were carried out in HepG2 cells treated with 100 µg per ml oxLDL for 24 to 48 h. 1-way or 2-way ANOVA was used for statistical analyses of datasets. RESULTS ER stress responses are elicited along all three arms of the unfolded protein response (UPR), with adverse cytotoxic and inflammatory responses in oxLDL-treated conditions. Interestingly, oxLDL-treated ER-stressed HepG2 cells manifested intriguingly low expression of BiP- the master regulator of ER stress, as observed earlier by various researchers in liver biopsies of Non-Alcoholic Steatohepatitis (NASH) patients. This study shows that overexpression of BiP rescues hepatic cells from cytotoxic and inflammatory mechanisms instigated by ER stress in combination with oxLDL, along the ER and mitochondrial membrane and restores cellular homeostasis. CONCLUSION The data provide interesting leads that identify patients with familial hypercholesterolemia conditions and potentially other Endoplasmic Reticulum Associated Degradation (ERAD) diseases as highly susceptible to developing hepatic insults with molecular signatures like those manifested in Non-Alcoholic Fatty Liver Disease (NAFLD) and NASH. LIMITATIONS AND FUTURE PERSPECTIVES Although the use of HepG2 cells as the model is a major caveat of the study, the findings of this research may be used as the pilot study to expand further investigations in primary hepatocytes or iPSC- derived cellular models.
Collapse
Affiliation(s)
- Divya Saro Varghese
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Deepu Oommen
- Present Address: Indian Institute of Science, C V Raman Road, 560012, Bangalore, India
| | - Anne John
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Bassam R Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.
- Zayed Centre for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
| |
Collapse
|
7
|
Miranda MA, Macias-Velasco JF, Schmidt H, Lawson HA. Integrated transcriptomics contrasts fatty acid metabolism with hypoxia response in β-cell subpopulations associated with glycemic control. BMC Genomics 2023; 24:156. [PMID: 36978008 PMCID: PMC10052828 DOI: 10.1186/s12864-023-09232-5] [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: 05/24/2022] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Understanding how heterogeneous β-cell function impacts diabetes is imperative for therapy development. Standard single-cell RNA sequencing analysis illuminates some factors driving heterogeneity, but new strategies are required to enhance information capture. RESULTS We integrate pancreatic islet single-cell and bulk RNA sequencing data to identify β-cell subpopulations based on gene expression and characterize genetic networks associated with β-cell function in obese SM/J mice. We identify β-cell subpopulations associated with basal insulin secretion, hypoxia response, cell polarity, and stress response. Network analysis associates fatty acid metabolism and basal insulin secretion with hyperglycemic-obesity, while expression of Pdyn and hypoxia response is associated with normoglycemic-obesity. CONCLUSIONS By integrating single-cell and bulk islet transcriptomes, our study explores β-cell heterogeneity and identifies novel subpopulations and genetic pathways associated with β-cell function in obesity.
Collapse
Affiliation(s)
- Mario A Miranda
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8232, Saint Louis, MO, 63110, USA
| | - Juan F Macias-Velasco
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8232, Saint Louis, MO, 63110, USA
| | - Heather Schmidt
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8232, Saint Louis, MO, 63110, USA
| | - Heather A Lawson
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8232, Saint Louis, MO, 63110, USA.
| |
Collapse
|
8
|
Kong Y, Niu A, Yuan W, Zhou Y, Xia M, Xiong X, Lu Y, Yin T, Zhang Y, Chen S, Huang Q, Zeng G, Huang Q. Interaction of FOXO1 and SUMOylated PPARγ1 induced by hyperlipidemia and hyperglycemia favors vascular endothelial insulin resistance and dysfunction. Vascul Pharmacol 2022; 147:107125. [PMID: 36252777 DOI: 10.1016/j.vph.2022.107125] [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: 04/21/2022] [Revised: 09/10/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
PPARγ1 and FOXO1 are the key transcription factors that regulate insulin sensitivity. We previously found that a small ubiquitin-related modifier of PPARγ1 at K77 (SUMOylation) favored endothelial insulin resistance (IR) induced by high fat/high glucose (HF/HG) administration. However, whether and how the crosstalk between SUMOylated PPARγ1 and FOXO1 would mediate the development of the endothelial IR and dysfunction remains unclear. Here, we emphasize how PPARγ1-K77 SUMOylation would interact with FOXO1 and participate in the development of the endothelial IR and dysfunction. Our results show that the combination of HF/HG and PPARγ1-K77 SUMOylation exhibits a synergistic deteriorative effect on the endothelial IR and dysfunction, presenting decreased NO levels and elevated ET-1 levels, weakened PI3K/Akt/eNOS signaling, and impaired endothelium-dependent vasodilation function. The further researches reveal that PPARγ1-K77 SUMOylation readily interacts with FOXO1, and FOXO1 occupies the PPAR response element (PPRE) which is supposed to be occupied by PPARγ, thus resulting in the decrease of PPARγ1 transcription activity and the mitigation of the PI3K/Akt signaling. Moreover, the mitigation of the PI3K/Akt signaling promotes in turn the accumulation of FOXO1 in the nucleus where FOXO1 interacts with the SUMOylated PPARγ1, thus exerting a positive feedback effect on IR pathogenesis. The findings uncover a novel association between PPARγ1-K77 SUMOylation and FOXO1, which contributes to our understanding of the pathogenesis of endothelial IR and dysfunction and provides novel pharmacological targets for diabetic angiopathy.
Collapse
Affiliation(s)
- Ying Kong
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China; Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Ailin Niu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Wanwan Yuan
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Yumeng Zhou
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Min Xia
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Xiaowei Xiong
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Yanli Lu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Tingting Yin
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Yanan Zhang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Sheng Chen
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Qianqian Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Guohua Zeng
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China.
| |
Collapse
|
9
|
Zou D, Yang P, Liu J, Dai F, Xiao Y, Zhao A, Huang N. Exosome-Loaded Pro-efferocytic Vascular Stent with Lp-PLA 2-Triggered Release for Preventing In-Stent Restenosis. ACS NANO 2022; 16:14925-14941. [PMID: 36066255 DOI: 10.1021/acsnano.2c05847] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The efferocytosis defect is regarded as a pivotal event of atherosclerosis. The failure to clear apoptotic cells in atherosclerotic plaques under vascular stents causes a failure to resolve the inflammation underneath. However, efferocytosis repair is still confined to nonstenting therapeutics. Here, we identified a pro-efferocytotic agent and accordingly developed a bioresponsive pro-efferocytotic vascular stent aimed for poststenting healing. Exosomes derived from mesenchymal stem cells were found to be able to regulate efferocytosis via SLC2a1, STAT3/RAC1, and CD300a pathways and modulate foam cell formation processes through a CD36-mediated pathway. Pro-efferocytotic exosomes were encapsulated into liposome-based multivesicular chambers and grafted onto vascular stents. The multivesicular vesicles were able to release exosomes under the Lp-PLA2 environment. Compared to bare metal stents, exosome-stents in the presence of Lp-PLA2 enhanced the ratio of apoptotic cell clearance and reduced the neointimal thickness in the mal-efferocytotic rat model. Overall, we identified a pro-efferocytic agent─exosomes that are able to regulate target cells via multiple signaling pathways and are good candidates to serve complex pathological environments, and this bioresponsive pro-efferocytotic vascular stent is an attractive approach for prevention of poststenting complications.
Collapse
Affiliation(s)
- Dan Zou
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Ping Yang
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Jianan Liu
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Fanfan Dai
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Yangyang Xiao
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Ansha Zhao
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Nan Huang
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| |
Collapse
|
10
|
3β-Hydroxy-5β-hydroxy-B-norcholestane-6β-carboxaldehyde (SEC-B) Induces Proinflammatory Activation of Human Endothelial Cells Associated with Nitric Oxide Production and Endothelial Nitric Oxide Synthase/Caveolin-1 Dysregulation. Antioxidants (Basel) 2022; 11:antiox11061148. [PMID: 35740044 PMCID: PMC9220016 DOI: 10.3390/antiox11061148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022] Open
Abstract
Oxysterols are a family of 27-carbon cholesterol oxidation derivatives found in low-density lipoproteins (LDLs) and atherosclerotic plaques where they trigger several biological responses involved in the initiation and progression of atherosclerosis. Several pieces of evidence suggest that oxysterols contribute to endothelial dysfunction (ED) due to their ability to alter membrane fluidity and cell permeability leading to inflammation, oxidative stress and apoptosis. The present study aimed to investigate the molecular events occurring in human microvascular endothelial cells (HMEC-1) in response to autoxidation-generated 3β-hydroxy-5β-hydroxy-B-norcholestane-6β-carboxaldehyde (SEC-B) exposure. Our results highlight that SEC-B rapidly activates HMEC-1 by inducing oxidative stress, nitric oxide (NO) production and pro-inflammatory cytokine release. Exposure to SEC-B up to 24 h results in persistent accumulation of the vasodilator NO paralleled by an upregulation of the endothelial nitric oxide synthase (eNOS) enzyme and downregulation of Caveolin-1 (Cav-1) protein levels. Moreover, reduced expression and extracellular release of the vasoconstrictor factor endothelin-1 (ET-1) are observed. Furthermore, SEC-B stimulates the expression of the cytokines interleukin-6 (IL-6) and tumor necrosis factor-like weak inducer of apoptosis (TWEAK). This proinflammatory state leads to increased monocyte recruitment on activated HMEC-1 cells. Our findings add new knowledge on the role of SEC-B in ED and further support its potential implication in atherosclerosis.
Collapse
|
11
|
Wang KY, Wang KJ, Ma Q. The expression and significance of p4E-BP1/4E-BP1 in prostate cancer. J Clin Lab Anal 2022; 36:e24332. [PMID: 35257419 PMCID: PMC8993610 DOI: 10.1002/jcla.24332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/17/2022] [Accepted: 02/19/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Although the phosphorylation of 4E-BP1 that has been detected in high-grade prostate cancer has been reported in previous studies, overexpression of p4E-BP1 and 4EBP1 and their clinical significance in prostate cancer still remain unknown. METHODS One hundred six samples of prostate tissues were collected and analyzed by immunohistochemistry with p4E-BP1 or 4E-BP1 specific antibodies. Everolimus was used to block the phosphorylation of p4E-BP1, and then flow cytometry, clone formation, transwell, and wound healing assays were performed to detect the survival and invasive ability of the prostate cancer cells. RESULTS We found that the expression of 4E-BP1 and p4E-BP1 was higher in prostate cancer tissues than in normal tissues. Interestingly, the expression of p4E-BP1 was significantly associated with Gleason score and lymph node metastasis, but had no obvious correlation with PSA and the presence of bone or visceral metastasis. However, no evident correlation was found between the positive expression of 4E-BP1 and these clinical characteristics. In in vitro experiments, we found similar results as the clinical presentation that 4E-BP1 and p4E-BP1 were low expressed in normal prostate epithelial cells, but in prostate cancer cells, as the malignancy increasing, 4E-BP1 and p4E-BP1 expression also gradually increased. Then, we used Everolimus to inhibit the phosphorylation of 4E-BP1 and found that Everolimus effectively reduced cloning formation, inhibited cell migration, and promoted apoptosis in a dose-dependent manner in PC3 cells. CONCLUSIONS These findings suggest that p4E-BP1 is a potential biomarker and therapy target for prostate cancer, and patients with high expressions of p4E-BP1 may benefit from Everolimus treatment.
Collapse
Affiliation(s)
- Kai-Yun Wang
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Ke-Jie Wang
- Translational Research Laboratory for Urology, the Key Laboratory of Ningbo City, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China.,Ningbo Clinical Research Center for Urological Disease, Ningbo, Zhejiang, China
| | - Qi Ma
- Translational Research Laboratory for Urology, the Key Laboratory of Ningbo City, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China.,Ningbo Clinical Research Center for Urological Disease, Ningbo, Zhejiang, China.,Comprehensive Urogenital Cancer Center, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
12
|
Zhang XZ, Fu L, Zou XY, Li S, Ma XD, Xie L, Pang B, Ma JB, Wang YJ, Du YR, Guo SC. Lung transcriptome analysis for the identification of genes involved in the hypoxic adaptation of plateau pika (Ochotona curzoniae). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2022; 41:100943. [PMID: 34861554 DOI: 10.1016/j.cbd.2021.100943] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/14/2021] [Accepted: 11/10/2021] [Indexed: 06/13/2023]
Abstract
The plateau pika, a typical hypoxia-tolerant mammal lives 3000-5000 m above sea level on the Qinghai-Tibet Plateau, has acquired many physiological and morphological characteristics and strategies in its adaptation to sustained, high-altitude hypoxia. Blunted hypoxic pulmonary vasoconstriction is one such strategy, but the genes involved in this strategy have not been elucidated. Here, we investigated the genes involved and their expression profiles in the lung transcriptome of plateau pikas subjected to different hypoxic conditions (using low-pressure oxygen cabins). A slight, right ventricular hypertrophy was observed in pikas of the control group (altitude: 3200 m) vs. those exposed to 5000 m altitude conditions for one week. Our assembly identified 67,774 genes; compared with their expression in the control animals, 866 and 8364 genes were co-upregulated and co-downregulated, respectively, in pikas subjected to 5000 m altitude conditions for 1 and 4 w. We elucidated pathways that were associated with pulmonary vascular arterial pressure, including vascular smooth muscle contraction, HIF-1 signalling, calcium signalling, cGMP-PKG signalling, and PI3K-Akt signalling based on the differentially expressed genes; the top-100 pathway enrichments were found between the control group and the group exposed to 5000 m altitude conditions for 4 w. The mRNA levels of 18 candidate gene showed that more than 83% of genes were expressed and the number of transcriptome The up-regulated genes were EPAS1, Hbα, iNOS, CX40, CD31, PPM1B, HIF-1α, MYLK, Pcdh12, Surfactant protein B, the down-regulated genes were RYR2, vWF, RASA1, CLASRP, HIF-3α. Our transcriptome data are a valuable resource for future genomic studies on plateau pika.
Collapse
Affiliation(s)
- Xu-Ze Zhang
- School of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; College of Ecological Environment and Resources, Qinghai Minzu University, Xining 810007, China; Key Laboratory of Evolution and Adaptation of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810001, China
| | - Lin Fu
- School of Life Science, Yunnan University, Yunnan 650091, China; Key Laboratory of Evolution and Adaptation of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810001, China
| | - Xiao-Yan Zou
- School of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Shuang Li
- Key Laboratory of Evolution and Adaptation of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810001, China
| | - Xiao-Dong Ma
- College of Ecological Environment and Resources, Qinghai Minzu University, Xining 810007, China; Key Laboratory of Evolution and Adaptation of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810001, China
| | - Ling Xie
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Bo Pang
- College of food science and biology, Hebei university of science and technology, Shijiazhuang 050018, China
| | - Jian-Bin Ma
- Key Laboratory of Biodiversity Formation Mechanism, Qinghai Normal University, Xining 810008, China
| | - Yu-Jun Wang
- Key Laboratory of Evolution and Adaptation of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810001, China
| | - Yu-Rong Du
- Key Laboratory of Biodiversity Formation Mechanism, Qinghai Normal University, Xining 810008, China.
| | - Song-Chang Guo
- School of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
13
|
Kianmehr A, Qujeq D, Bagheri A, Mahrooz A. Oxidized LDL-regulated microRNAs for evaluating vascular endothelial function: molecular mechanisms and potential biomarker roles in atherosclerosis. Crit Rev Clin Lab Sci 2021; 59:40-53. [PMID: 34523391 DOI: 10.1080/10408363.2021.1974334] [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] [Indexed: 01/20/2023]
Abstract
As a simple monolayer, vascular endothelial cells can respond to physicochemical stimuli. In addition to promoting the formation of foam cells, oxidized low-density lipoprotein (ox-LDL) contributes to the atherosclerotic process through different mechanisms, including endothelial cell dysfunction. As conserved noncoding RNAs, microRNAs (miRNAs) naturally lie in different genomic positions and post-transcriptionally regulate the expression of many genes. They participate in integrated networks formed under stress to maintain cellular homeostasis, vascular inflammation, and metabolism. These small RNAs constitute therapeutic targets in different diseases, including atherosclerosis, and their role as biomarkers is crucial given their detectability even years before the emergence of diseases. This review was performed to investigate the role of ox-LDL-regulated miRNAs in atherosclerosis, their molecular mechanisms, and their application as biomarkers of vascular endothelial cell dysfunction.
Collapse
Affiliation(s)
- Anvarsadat Kianmehr
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abouzar Bagheri
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdolkarim Mahrooz
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
14
|
Lysophosphatidylcholine in phospholipase A 2-modified LDL triggers secretion of angiopoietin 2. Atherosclerosis 2021; 327:87-99. [PMID: 34020784 DOI: 10.1016/j.atherosclerosis.2021.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND AIMS Secretory phospholipase A2 (PLA2) hydrolyzes LDL phospholipids generating modified LDL particles (PLA2-LDL) with increased atherogenic properties. Exocytosis of Weibel-Palade bodies (WPB) releases angiopoietin 2 (Ang2) and externalizes P-selectin, which both play important roles in vascular inflammation. Here, we investigated the effects of PLA2-LDL on exocytosis of WPBs. METHODS Human coronary artery endothelial cells (HCAECs) were stimulated with PLA2- LDL, and its uptake and effect on Ang2 release, leukocyte adhesion, and intracellular calcium levels were measured. The effects of PLA2-LDL on Ang2 release and WPB exocytosis were measured in and ex vivo in mice. RESULTS Exposure of HCAECs to PLA2-LDL triggered Ang2 secretion and promoted leukocyte-HCAEC interaction. Lysophosphatidylcholine was identified as a critical component of PLA2-LDL regulating the WPB exocytosis, which was mediated by cell-surface proteoglycans, phospholipase C, intracellular calcium, and cytoskeletal remodeling. PLA2-LDL also induced murine endothelial WPB exocytosis in blood vessels in and ex vivo, as evidenced by secretion of Ang2 in vivo, P-selectin translocation to plasma membrane in intact endothelial cells in thoracic artery and tracheal vessels, and reduced Ang2 staining in tracheal endothelial cells. Finally, in contrast to normal human coronary arteries, in which Ang2 was present only in the endothelial layer, at sites of advanced atherosclerotic lesions, Ang2 was detected also in the intima, media, and adventitia. CONCLUSIONS Our studies reveal PLA2-LDL as a potent agonist of endothelial WPB exocytosis, resulting in increased secretion of Ang2 and translocation of P-selectin. The results provide mechanistic insight into PLA2-LDL-dependent promotion of vascular inflammation and atherosclerosis.
Collapse
|
15
|
Roldán Gallardo FF, Quintar AA. The pathological growth of the prostate gland in atherogenic contexts. Exp Gerontol 2021; 148:111304. [PMID: 33676974 DOI: 10.1016/j.exger.2021.111304] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 02/06/2023]
Abstract
The human prostate is an androgen-dependent gland where an imbalance in cell proliferation can lead to benign prostatic hyperplasia (BPH), which results in voiding lower urinary tract symptoms in the elderly. In the last decades, novel evidence has suggested that BPH might represent an element into the wide spectrum of disorders conforming the Metabolic Syndrome (MS). The dyslipidemic state and the other atherogenic factors of the MS have been shown to induce, maintain and/or aggravate the pathological growth of different organs, with data regarding the prostate being still limited. We here review the available epidemiological and experimental studies about the association of BPH with dyslipidemias. In particular, we have focused on Oxidized Low-Density Lipoproteins (OxLDL) as a potential trigger for vascular disease and cellular proliferation in atherogenic contexts, analyzing their putative molecular mechanisms, including the induction of specific extracellular vesicles (EVs)-derived miRNAs. In addition to the epidemiological evidence, OxLDL is proposed to play a fundamental role in the upregulation of prostatic cell proliferation by activating the Rho/Akt/p27Kip1 pathway in atherogenic contexts. miR-21, miR-141, miR-143, miR-145, miR-155, and miR-221 would be involved in the transcription of genes related to the proliferative process. Although much remains to be investigated regarding the impact of OxLDL, its receptors, and molecular mechanisms on the prostate, it is clear that EVs and miRNAs represent a promising target for proliferative pathologies of the prostate gland.
Collapse
Affiliation(s)
- Franco F Roldán Gallardo
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Amado A Quintar
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina.
| |
Collapse
|
16
|
Weighed Gene Coexpression Network Analysis Screens the Potential Long Noncoding RNAs and Genes Associated with Progression of Coronary Artery Disease. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2020; 2020:8183420. [PMID: 32695216 PMCID: PMC7361886 DOI: 10.1155/2020/8183420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/17/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022]
Abstract
Background Coronary artery disease (CAD) is a type of heart disease with a high morbidity rate. This study is aimed at identifying potential biomarkers closely related to the progression of CAD. Materials and Methods A microarray dataset of GSE59867 was downloaded from a public database, Gene Expression Omnibus, which included 46 cases of stable CAD without a history of myocardial infarction (MI), 30 cases of MI without heart failure (HF), and 34 cases of MI with HF. Differentially expressed long noncoding RNAs (DElncRNAs) and mRNAs (DEmRNAs) were identified by the limma package, and functions of DEmRNAs were annotated by Gene Ontology and KEGG pathways. In addition, weighed gene coexpression network analysis (WGCNA) was used to construct a coexpression network of DEmRNAs, and a disease-related lncRNAs-mRNAs-pathway network was constructed. Finally, the datasets of GSE61145 and GSE57338 were used to verify the expression levels of the above highly correlated candidates. Results A total of 2362 upregulated mRNAs and 2816 downregulated mRNAs, as well as 235 upregulated lncRNAs and 113 downregulated lncRNAs were screened. These genes were significantly enriched in “cytokine-cytokine receptor interaction,” “RIG-I-like receptor signaling pathway,” and “natural killer cell-mediated cytotoxicity.” Five modules including 1201 DEmRNAs were enriched in WGCNA. A coexpression network including 19 DElncRNAs and 413 DEmRNAs was constructed. These genes were significantly enriched in “phosphatidylinositol signaling system,” “insulin signaling pathway,” and “MAPK signaling pathway”. Disease-related gene-pathway network suggested FASN in “insulin signaling pathway,” DGKZ in “phosphatidylinositol signaling system,” and TNFRSF1A in “MAPK signaling pathway” were involved in MI. Conclusion FASN, DGKZ, and TNFRSF1A were revealed to be CAD progression-associated genes by WGCNA coexpression network analysis.
Collapse
|
17
|
Wu T, Xu W, Wang Y, Tao M, Hu Z, Lv B, Hui Y, Du H. OxLDL enhances choroidal neovascularization lesion through inducing vascular endothelium to mesenchymal transition process and angiogenic factor expression. Cell Signal 2020; 70:109571. [DOI: 10.1016/j.cellsig.2020.109571] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/16/2020] [Accepted: 02/16/2020] [Indexed: 10/25/2022]
|
18
|
Chen Q, Li X, Kong L, Xu Q, Wang Z, Lv Q. miR-101-3p induces vascular endothelial cell dysfunction by targeting tet methylcytosine dioxygenase 2. Acta Biochim Biophys Sin (Shanghai) 2020; 52:180-191. [PMID: 31990036 DOI: 10.1093/abbs/gmz154] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 12/27/2022] Open
Abstract
Endothelial cell (EC) dysfunction represents an early key event in atherosclerosis. Recently, MicroRNAs have been demonstrated to regulate EC function. miR-101-3p has been discovered to regulate cell apoptosis and proliferation in cardiovascular diseases. Therefore, the aim of the current study was to clarify whether miR-101-3p regulates the dysfunction of vascular endothelial cells. In this study, the transfection of human umbilical vein endothelial cells (HUVECs) with miR-101-3p mimic induced reactive oxygen species (ROS) production, EC dysfunction, and activated nuclear factor-κB (NF-κB), whereas transfection with miR-101-3p inhibitor alleviated these events. The antioxidant N-acetylcysteine alleviated miR-101-3p-induced EC dysfunction. Moreover, we observed that miR-101-3p inhibited the expression of tet methylcytosine dioxygenase 2 (TET2) at the posttranscriptional level, resulting in increased ROS production and activated NF-κB. TET2 overexpression inhibited ROS production, EC dysfunction, and NF-κB activation in miR-101-3p-transfected HUVECs. These results indicate that miR-101-3p induces EC dysfunction by targeting TET2, which regulates ROS production, EC dysfunction, and NF-κB activation. Taken together, our current study reveals a novel pathway associated with EC dysfunction. The modulation of miR-101-3p and TET2 expression levels may serve as a potential target for therapeutic strategies for atherosclerosis.
Collapse
Affiliation(s)
- Qiaoli Chen
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaoye Li
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lingjun Kong
- Department of Clincial Pharmacy, Obstetrics & Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Qing Xu
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zi Wang
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qianzhou Lv
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
19
|
Bogachkov YY, Chen L, Le Master E, Fancher IS, Zhao Y, Aguilar V, Oh MJ, Wary KK, DiPietro LA, Levitan I. LDL induces cholesterol loading and inhibits endothelial proliferation and angiogenesis in Matrigels: correlation with impaired angiogenesis during wound healing. Am J Physiol Cell Physiol 2020; 318:C762-C776. [PMID: 31995410 DOI: 10.1152/ajpcell.00495.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Hypercholesterolemia is a major risk factor for adverse cardiovascular outcomes, but its effect on angiogenesis and wound healing is not well understood. In this study, using a combination of mass spectrometry and laurdan two-photon imaging, we show that elevated levels of low-density lipoprotein (LDL), like those seen in hypercholesterolemic patients, lead to an increase in both free cholesterol and cholesterol esters, as well as increase in lipid order of endothelial cell membranes. Notably, these effects are distinct and opposite to the lack of cholesterol loading and the disruption of lipid order observed in our earlier studies in response to oxidized LDL (oxLDL). The same pathological level of LDL leads to a significant inhibition of endothelial proliferation and cell cycle arrest in G2/M phase, whereas oxLDL enhances endothelial proliferation in S phase of the cycle. LDL but not oxLDL suppresses the expression of vascular endothelial growth factor receptor-2 while enhancing the expression of vascular endothelial growth factor (VEGF). Furthermore, we show that aged (8-10 mo) hypercholesterolemic apolipoprotein E-deficient (ApoE-/-) mice display delayed wound closure compared with age-matched C57/BL6 wild-type controls following a skin punch biopsy. The delay in wound healing is associated with a decreased expression of cluster of differentiation 31 platelet endothelial cell adhesion molecule endothelial marker and decreased angiogenesis within the wound bed. Furthermore, decreased endothelial responsiveness to the growth factors VEGF and basic fibroblast growth factor is observed in ApoE-/- mice in Matrigel plugs and in Matrigels with high levels of LDL in wild-type mice. We propose that plasma hypercholesterolemia is antiangiogenic due to elevated levels of LDL.
Collapse
Affiliation(s)
- Yedida Y Bogachkov
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois.,Department of Cellular and Molecular Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois
| | - Elizabeth Le Master
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Ibra S Fancher
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Yan Zhao
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois
| | - Victor Aguilar
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Myung-Jin Oh
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Kishore K Wary
- Department of Cellular and Molecular Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Luisa A DiPietro
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois
| | - Irena Levitan
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois.,Department of Cellular and Molecular Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
20
|
Hong FF, Liang XY, Liu W, Lv S, He SJ, Kuang HB, Yang SL. Roles of eNOS in atherosclerosis treatment. Inflamm Res 2019; 68:429-441. [DOI: 10.1007/s00011-019-01229-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 03/16/2019] [Accepted: 03/18/2019] [Indexed: 02/04/2023] Open
|
21
|
Tetramethylpyrazine and Paeoniflorin Inhibit Oxidized LDL-Induced Angiogenesis in Human Umbilical Vein Endothelial Cells via VEGF and Notch Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:3082507. [PMID: 30584451 PMCID: PMC6280302 DOI: 10.1155/2018/3082507] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/06/2018] [Accepted: 11/07/2018] [Indexed: 02/01/2023]
Abstract
Atherosclerotic plaque angiogenesis is key factor in plaque instability and vulnerability, and low concentrations of oxidized low density lipoprotein (ox-LDL) promote the in vitro angiogenesis of endothelial cells and play an important role in plaque angiogenesis. Ligusticum chuanxiong Hort. and Radix Paeoniae Rubra herb pair in Chinese medicine obtains the optimum therapeutic efficacy in atherosclerosis, and their major active ingredients tetramethylpyrazine (TMP) and paeoniflorin (PF) are reported to alleviate atherosclerosis. The aim of this study was to investigate the effects of TMP and PF on ox-LDL-induced angiogenesis and the underlying mechanism. Human umbilical vein endothelial cells (HUVECs) were incubated with ox-LDL and were then treated with TMP, PF, or a combination of TMP and PF. Cell proliferation, migration, tube formation, and the expression of angiogenesis-related proteins were measured. Synergism was evaluated using the combination index in cell proliferation. We found that TMP and PF attenuated the in vitro angiogenesis in ox-LDL-induced HUVECs. In addition, the combination of TMP and PF not only inhibited the ox-LDL-induced expression of CD31, vascular endothelial growth factor (VEGF), and VEGF receptor 2 (VEGFR2) but also decreased the ox-LDL-induced expression of Notch1, Jagged1, and Hes1. In summary, the combination of TMP and PF suppresses ox-LDL-induced angiogenesis in HUVECs by inhibiting both the VEGF/VEGFR2 and the Jagged1/Notch1 signaling pathways, which might contribute to the stability of plaques in atherosclerosis.
Collapse
|
22
|
Willson C, Watanabe M, Tsuji-Hosokawa A, Makino A. Pulmonary vascular dysfunction in metabolic syndrome. J Physiol 2018; 597:1121-1141. [PMID: 30125956 DOI: 10.1113/jp275856] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/30/2018] [Indexed: 12/20/2022] Open
Abstract
Metabolic syndrome is a critically important precursor to the onset of many diseases, such as cardiovascular disease, and cardiovascular disease is the leading cause of death worldwide. The primary risk factors of metabolic syndrome include hyperglycaemia, abdominal obesity, dyslipidaemia, and high blood pressure. It has been well documented that metabolic syndrome alters vascular endothelial and smooth muscle cell functions in the heart, brain, kidney and peripheral vessels. However, there is less information available regarding how metabolic syndrome can affect pulmonary vascular function and ultimately increase an individual's risk of developing various pulmonary vascular diseases, such as pulmonary hypertension. Here, we review in detail how metabolic syndrome affects pulmonary vascular function.
Collapse
Affiliation(s)
- Conor Willson
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Makiko Watanabe
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | | | - Ayako Makino
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
23
|
Qian Y, Song J, Zhao X, Chen W, Ouyang Y, Yuan W, Fan C. 3D Fabrication with Integration Molding of a Graphene Oxide/Polycaprolactone Nanoscaffold for Neurite Regeneration and Angiogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1700499. [PMID: 29721407 PMCID: PMC5908351 DOI: 10.1002/advs.201700499] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 12/02/2017] [Indexed: 05/17/2023]
Abstract
Treating peripheral nerve injury faces major challenges and may benefit from bioactive scaffolds due to the limited autograft resources. Graphene oxide (GO) has emerged as a promising nanomaterial with excellent physical and chemical properties. GO has functional groups that confer biocompatibility that is better than that of graphene. Here, GO/polycaprolactone (PCL) nanoscaffolds are fabricated using an integration molding method. The nanoscaffolds exhibit many merits, including even GO nanoparticle distribution, macroporous structure, and strong mechanical support. Additionally, the process enables excellent quality control. In vitro studies confirm the advantages of the GO/PCL nanoscaffolds in terms of Schwann cell proliferation, viability, and attachment, as well as neural characteristics maintenance. This is the first study to evaluate the in vivo performance of GO-based nanoscaffolds in this context. GO release and PCL biodegradation is analyzed after long-term in vivo study. It is also found that the GO/PCL nerve guidance conduit could successfully repair a 15 mm sciatic nerve defect. The pro-angiogenic characteristic of GO is evaluated in vivo using immunohistochemistry. In addition, the AKT-endothelial nitric oxide synthase (eNOS)-vascular endothelial growth factor (VEGF) signaling pathway might play a major role in the angiogenic process. These findings demonstrate that the GO/PCL nanoscaffold efficiently promotes functional and morphological recovery in peripheral nerve regeneration, indicating its promise for tissue engineering applications.
Collapse
Affiliation(s)
- Yun Qian
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital600 Yishan RoadShanghai200233China
- Shanghai Sixth People's Hospital East CampusShanghai University of Medicine and HealthShanghai201306China
| | - Jialin Song
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital600 Yishan RoadShanghai200233China
| | - Xiaotian Zhao
- School of PharmacyShanghai Jiao Tong UniversityNo. 800 Dongchuan RoadShanghai200240China
| | - Wei Chen
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital600 Yishan RoadShanghai200233China
| | - Yuanming Ouyang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital600 Yishan RoadShanghai200233China
- Shanghai Sixth People's Hospital East CampusShanghai University of Medicine and HealthShanghai201306China
| | - Weien Yuan
- School of PharmacyShanghai Jiao Tong UniversityNo. 800 Dongchuan RoadShanghai200240China
| | - Cunyi Fan
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital600 Yishan RoadShanghai200233China
| |
Collapse
|
24
|
Bao MH, Li GY, Huang XS, Tang L, Dong LP, Li JM. Long Noncoding RNA LINC00657 Acting as a miR-590-3p Sponge to Facilitate Low Concentration Oxidized Low-Density Lipoprotein–Induced Angiogenesis. Mol Pharmacol 2018; 93:368-375. [DOI: 10.1124/mol.117.110650] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/29/2018] [Indexed: 12/30/2022] Open
|
25
|
Dobrynina LA, Gnedovskaya EV, Shabalina AA, Sergeeva AN, Kravchenko MA, Nikolaeva NS. Biomarkers and mechanisms of early vascular damage. Zh Nevrol Psikhiatr Im S S Korsakova 2018; 118:23-32. [DOI: 10.17116/jnevro201811812223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
26
|
Chen JY, Ye ZX, Wang XF, Chang J, Yang MW, Zhong HH, Hong FF, Yang SL. Nitric oxide bioavailability dysfunction involves in atherosclerosis. Biomed Pharmacother 2017; 97:423-428. [PMID: 29091892 DOI: 10.1016/j.biopha.2017.10.122] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/22/2017] [Accepted: 10/22/2017] [Indexed: 12/25/2022] Open
Abstract
The pathological characteristics of atherosclerosis (AS) include lipid accumulation, fibrosis formation and atherosclerotic plaque produced in artery intima, which leads to vascular sclerosis, lumen stenosis and irritates the ischemic changes of corresponding organs. Endothelial dysfunction was closely associated with AS. Nitric oxide (NO) is a multifunctional signaling molecule involved in the maintenance of metabolic and cardiovascular homeostasis. NO is also a potent endogenous vasodilator and enters for the key processes that suppresses the formation vascular lesion even AS. NO bioavailability indicates the production and utilization of endothelial NO in organisms, its decrease is related to oxidative stress, lipid infiltration, the expressions of some inflammatory factors and the alteration of vascular tone, which plays an important role in endothelial dysfunction. The enhancement of arginase activity and the increase in asymmetric dimethylarginine and hyperhomocysteinemia levels all contribute to AS by intervening NO bioavailability in human beings. Diabetes mellitus, obesity, chronic kidney disease and smoking, etc., also participate in AS by influencing NO bioavailability and NO level. Here, we reviewed the relationship between NO bioavailability and AS according the newest literatures.
Collapse
Affiliation(s)
- Jing-Yi Chen
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Zi-Xin Ye
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Xiu-Fen Wang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Jian Chang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Mei-Wen Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Hua-Hua Zhong
- Department of Experimental Teaching Center, Nanchang University, Nanchang 330031, China
| | - Fen-Fang Hong
- Department of Experimental Teaching Center, Nanchang University, Nanchang 330031, China.
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
27
|
Shi X, Zhou K, Huang F, Wang C. Interaction of hydroxyapatite nanoparticles with endothelial cells: internalization and inhibition of angiogenesis in vitro through the PI3K/Akt pathway. Int J Nanomedicine 2017; 12:5781-5795. [PMID: 28848353 PMCID: PMC5557617 DOI: 10.2147/ijn.s140179] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Nano-hydroxyapatite (nano-HAP) has been proposed as a better candidate for bone tissue engineering; however, the interactions of nano-HAP with endothelial cells are currently unclear. In this study, HAP nanoparticles (HANPs; 20 nm np20 and 80 nm np80) and micro-sized HAP particles (m-HAP; 12 μm) were employed to explore and characterize cellular internalization, subcellular distribution, effects of HANPs on endothelial cell function and underlying mechanisms using human umbilical vein endothelial cells (HUVECs) as an in vitro model. It was found that HANPs were able to accumulate in the cytoplasm, and both adhesion and uptake of the HANPs followed a function of time; compared to np80, more np20 had been uptaken at the end of the observation period. HANPs were mainly uptaken via clathrin- and caveolin-mediated endocytosis, while macropinocytosis was the main pathway for m-HAP uptake. Unexpectedly, exposure to HANPs suppressed the angiogenic ability of HUVECs in terms of cell viability, cell cycle, apoptosis response, migration and capillary-like tube formation. Strikingly, HANPs reduced the synthesis of nitric oxide (NO) in HUVECs, which was associated with the inhibition of phosphatidylinositol 3-kinase (PI3K) and phosphorylation of eNOS. These findings provide additional insights into specific biological responses as HANPs interface with endothelial cells.
Collapse
Affiliation(s)
- Xingxing Shi
- Department of Prosthodontics, Jiangsu Key Laboratory of Oral Diseases, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Kai Zhou
- Department of Prosthodontics, Jiangsu Key Laboratory of Oral Diseases, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Fei Huang
- Department of Prosthodontics, Jiangsu Key Laboratory of Oral Diseases, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Chen Wang
- Department of Prosthodontics, Jiangsu Key Laboratory of Oral Diseases, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
28
|
Zhang C, Adamos C, Oh MJ, Baruah J, Ayee MAA, Mehta D, Wary KK, Levitan I. oxLDL induces endothelial cell proliferation via Rho/ROCK/Akt/p27 kip1 signaling: opposite effects of oxLDL and cholesterol loading. Am J Physiol Cell Physiol 2017; 313:C340-C351. [PMID: 28701359 DOI: 10.1152/ajpcell.00249.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 07/05/2017] [Accepted: 07/05/2017] [Indexed: 12/19/2022]
Abstract
Oxidized modifications of LDL (oxLDL) play a key role in the development of endothelial dysfunction and atherosclerosis. However, the underlying mechanisms of oxLDL-mediated cellular behavior are not completely understood. Here, we compared the effects of two major types of oxLDL, copper-oxidized LDL (Cu2+-oxLDL) and lipoxygenase-oxidized LDL (LPO-oxLDL), on proliferation of human aortic endothelial cells (HAECs). Cu2+-oxLDL enhanced HAECs' proliferation in a dose- and degree of oxidation-dependent manner. Similarly, LPO-oxLDL also enhanced HAEC proliferation. Mechanistically, both Cu2+-oxLDL and LPO-oxLDL enhance HAEC proliferation via activation of Rho, Akt phosphorylation, and a decrease in the expression of cyclin-dependent kinase inhibitor 1B (p27kip1). Both Cu2+-oxLDL or LPO-oxLDL significantly increased Akt phosphorylation, whereas an Akt inhibitor, MK2206, blocked oxLDL-induced increase in HAEC proliferation. Blocking Rho with C3 or its downstream target ROCK with Y27632 significantly inhibited oxLDL-induced Akt phosphorylation and proliferation mediated by both Cu2+- and LPO-oxLDL. Activation of RhoA was blocked by Rho-GDI-1, which also abrogated oxLDL-induced Akt phosphorylation and HAEC proliferation. In contrast, blocking Rac1 in these cells had no effect on oxLDL-induced Akt phosphorylation or cell proliferation. Moreover, oxLDL-induced Rho/Akt signaling downregulated cell cycle inhibitor p27kip1 Preloading these cells with cholesterol, however, prevented oxLDL-induced Akt phosphorylation and HAEC proliferation. These findings provide a new understanding of the effects of oxLDL on endothelial proliferation, which is essential for developing new treatments against neovascularization and progression of atherosclerosis.
Collapse
Affiliation(s)
- Chongxu Zhang
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Crystal Adamos
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Myung-Jin Oh
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Jugajyoti Baruah
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Manuela A A Ayee
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Dolly Mehta
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Kishore K Wary
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Irena Levitan
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| |
Collapse
|
29
|
Fan ZM, Wang DY, Yang JM, Lin ZX, Lin YX, Yang AL, Fan H, Cao M, Yuan SY, Liu ZJ, Zhou X, Wang YH. Dalbergia odorifera extract promotes angiogenesis through upregulation of VEGFRs and PI3K/MAPK signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2017; 204:132-141. [PMID: 28412217 DOI: 10.1016/j.jep.2017.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/21/2017] [Accepted: 04/07/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The heart wood of Dalbergia odorifera is a Chinese herbal medicine commonly used for the treatment of various ischemic diseases in Chinese medicine practice. AIM OF THE STUDY In this study, therapeutic angiogenesis effects of the Dalbergia odorifera extract (DOE) were investigated on transgenic zebrafish in vivo and human umbilical vein endothelial cells (HUVECs) in vitro. MATERIALS AND METHODS The pro-angiogenic effects of DOE on zebrafish were examined by subintestinal vessels (SIVs) sprouting assay and intersegmental vessels (ISVs) injury assay. And the pro-angiogenic effects of DOE on HUVECs were examined by MTT, scratch assay, protein chip and western blot. RESULTS In the in vivo studies, we found that DOE was able to dose-dependently promote angiogenesis in zebrafish SIVs area. In addition, DOE could also restore the injury in zebrafish ISVs area and upregulate the reduced mRNA expression of VEGFRs including kdr, kdrl and flt-1 induced by VEGF receptor kinase inhibitor II (VRI). In the in vitro studies, we observed that DOE promoted the proliferation, migration of HUVECs and also restored the injury induced by VRI. Moreover, protein chip and western blot experiments showed the PI3K/MAPK cell proliferation/migration pathway were activated by DOE. CONCLUSIONS DOE has a therapeutic effects on angiogenesis, and its mechanism may be related to adjusting the VEGFRs mRNA and activation of PI3K/MAPK signaling pathway. These results suggest a strong potential for Dalbergia odorifera to be developed as an angiogenesis-promoting therapeutic.
Collapse
Affiliation(s)
- Zhu-Ming Fan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Da-Ying Wang
- Central Hospital of Shanghai Xuhui District, Shanghai 200031, PR China
| | - Jian-Mei Yang
- Central Hospital of Shanghai Putuo District, Shanghai 200062, PR China
| | - Zhi-Xiu Lin
- Faculty of Science, School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Yun-Xiao Lin
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Ai-Lin Yang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Hua Fan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Min Cao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Su-Yun Yuan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Zong-Jun Liu
- Central Hospital of Shanghai Xuhui District, Shanghai 200031, PR China
| | - Xin Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China.
| | - You-Hua Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China.
| |
Collapse
|
30
|
4-Hydroxynonenal Contributes to Angiogenesis through a Redox-Dependent Sphingolipid Pathway: Prevention by Hydralazine Derivatives. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9172741. [PMID: 28479957 PMCID: PMC5396448 DOI: 10.1155/2017/9172741] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/01/2017] [Indexed: 12/23/2022]
Abstract
The neovascularization of atherosclerotic lesions is involved in plaque development and may contribute to intraplaque hemorrhage and plaque fragilization and rupture. Among the various proangiogenic agents involved in the neovascularization process, proatherogenic oxidized LDLs (oxLDLs) contribute to the formation of tubes via the generation of sphingosine 1-phosphate (S1P), a major mitogenic and proangiogenic sphingolipid mediator. In this study, we investigated whether 4-hydroxynonenal (4-HNE), an aldehydic lipid oxidation product abundantly present in oxLDLs, contributes to their proangiogenic properties. Immunofluorescence analysis of human atherosclerotic lesions from carotid endarterectomy showed the colocalization of HNE-adducts with CD31, a marker of endothelial cells, suggesting a close relationship between 4-HNE and neovessel formation. In vitro, low 4-HNE concentration (0.5-1 µM) elicited the formation of tubes by human microvascular endothelial cells (HMEC-1), whereas higher concentrations were not angiogenic. The formation of tubes by 4-HNE involved the generation of reactive oxygen species and the activation of the sphingolipid pathway, namely, the neutral type 2 sphingomyelinase and sphingosine kinase-1 (nSMase2/SK-1) pathway, indicating a role for S1P in the angiogenic signaling of 4-HNE. Carbonyl scavengers hydralazine and bisvanillyl-hydralazone inhibited the nSMase2/SK1 pathway activation and the formation of tubes on Matrigel® evoked by 4-HNE. Altogether, these results emphasize the role of 4-HNE in the angiogenic effect of oxLDLs and point out the potential interest of pharmacological carbonyl scavengers to prevent the neovascularization process.
Collapse
|
31
|
Li JB, Wang HY, Yao Y, Sun QF, Liu ZH, Liu SQ, Zhuang JL, Wang YP, Liu HY. Overexpression of microRNA-138 alleviates human coronary artery endothelial cell injury and inflammatory response by inhibiting the PI3K/Akt/eNOS pathway. J Cell Mol Med 2017; 21:1482-1491. [PMID: 28371277 PMCID: PMC5542903 DOI: 10.1111/jcmm.13074] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/29/2016] [Indexed: 12/24/2022] Open
Abstract
This study aimed to investigate the role of miR‐138 in human coronary artery endothelial cell (HCAEC) injury and inflammatory response and the involvement of the PI3K/Akt/eNOS signalling pathway. Oxidized low‐density lipoprotein (OX‐LDL)‐induced HCAEC injury models were established and assigned to blank, miR‐138 mimic, miR‐138 inhibitor, LY294002 (an inhibitor of the PI3K/Akt/eNOS pathway), miR‐138 inhibitor + LY294002 and negative control (NC) groups. qRT‐PCR and Western blotting were performed to detect the miR‐138, PI3K, Akt and eNOS levels and the protein expressions of PI3K, Akt, eNOS, p‐Akt, p‐eNOS, Bcl‐2, Bax and caspase‐3. ELISAs were employed to measure the expressions of TNF‐α, IL‐4, IL‐6, IL‐8, IL‐10 and nitric oxide (NO) and the activities of lactate dehydrogenase (LDH) and eNOS. MTT and flow cytometry were performed to assess the proliferation and apoptosis of HCAECs. Compared to the blank group, PI3K, Akt and eNOS were down‐regulated in the miR‐138 mimic and LY294002 groups but were up‐regulated in the miR‐138 inhibitor group. The miR‐138 mimic and LY294002 groups showed decreased concentrations of TNF‐α, IL‐6, IL‐8 and NO and reduced activities of LDH and eNOS, while opposite trends were observed in the miR‐138 inhibitor group. The concentrations of IL‐4 and IL‐10 increased in the miR‐138 mimic and LY294002 groups but decreased in the miR‐138 inhibitor group. The miR‐138 mimic and LY294002 groups had significantly decreased cell proliferation and increased cell apoptosis compared to the blank group. These findings indicate that up‐regulation of miR‐138 alleviates HCAEC injury and inflammatory response by inhibiting the PI3K/Akt/eNOS signalling pathway.
Collapse
Affiliation(s)
- Jing-Bo Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hai-Yang Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ye Yao
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qing-Feng Sun
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zong-Hong Liu
- Department of Cardiac Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Si-Qi Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jun-Li Zhuang
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yun-Peng Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hong-Yu Liu
- Department of Cardiac Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
32
|
Camaré C, Pucelle M, Nègre-Salvayre A, Salvayre R. Angiogenesis in the atherosclerotic plaque. Redox Biol 2017; 12:18-34. [PMID: 28212521 PMCID: PMC5312547 DOI: 10.1016/j.redox.2017.01.007] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a multifocal alteration of the vascular wall of medium and large arteries characterized by a local accumulation of cholesterol and non-resolving inflammation. Atherothrombotic complications are the leading cause of disability and mortality in western countries. Neovascularization in atherosclerotic lesions plays a major role in plaque growth and instability. The angiogenic process is mediated by classical angiogenic factors and by additional factors specific to atherosclerotic angiogenesis. In addition to its role in plaque progression, neovascularization may take part in plaque destabilization and thromboembolic events. Anti-angiogenic agents are effective to reduce atherosclerosis progression in various animal models. However, clinical trials with anti-angiogenic drugs, mainly anti-VEGF/VEGFR, used in anti-cancer therapy show cardiovascular adverse effects, and require additional investigations.
Collapse
Affiliation(s)
- Caroline Camaré
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France; Université Paul Sabatier Toulouse III, Faculty of Medicine, Biochemistry Departement, Toulouse, France; CHU Toulouse, Rangueil, 1 avenue Jean Poulhès, TSA 50032, 31059 Toulouse Cedex 9, France
| | - Mélanie Pucelle
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France
| | - Anne Nègre-Salvayre
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France.
| | - Robert Salvayre
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France; Université Paul Sabatier Toulouse III, Faculty of Medicine, Biochemistry Departement, Toulouse, France; CHU Toulouse, Rangueil, 1 avenue Jean Poulhès, TSA 50032, 31059 Toulouse Cedex 9, France.
| |
Collapse
|
33
|
Camaré C, Augé N, Pucelle M, Saint-Lebes B, Grazide MH, Nègre-Salvayre A, Salvayre R. The neutral sphingomyelinase-2 is involved in angiogenic signaling triggered by oxidized LDL. Free Radic Biol Med 2016; 93:204-16. [PMID: 26855418 DOI: 10.1016/j.freeradbiomed.2016.02.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/31/2016] [Accepted: 02/02/2016] [Indexed: 12/31/2022]
Abstract
Capillaries of the external part of the normal arterial wall constitute the vasa vasorum network. In atherosclerotic lesions, neovascularization occurs in areas of intimal hyperplasia where it may promote plaque expansion, and intraplaque hemorrhage. Oxidized LDL that are present in atherosclerotic areas activate various angiogenic signaling pathways, including reactive oxygen species and the sphingosine kinase/sphingosine-1-phosphate pathway. We aimed to investigate whether oxidized LDL-induced angiogenesis requires neutral sphingomyelinase-2 activation and the neutral sphingomyelinase-2/sphingosine kinase-1 pathway. The role of neutral sphingomyelinase-2 in angiogenic signaling was investigated in Human Microvascular Endothelial Cells (HMEC-1) forming capillary tube on Matrigel and in vivo in the Matrigel plug assay in C57BL/6 mice and in the chicken chorioallantoic membrane model. Low concentration of human oxidized LDL elicits HMEC-1 capillary tube formation and neutral sphingomyelinase-2 activation, which were blocked by neutral sphingomyelinase-2 inhibitors, GW4869 and specific siRNA. This angiogenic effect was mimicked by low concentration of C6-Ceramide and was inhibited by sphingosine kinase-1 inhibitors. Upstream of neutral sphingomyelinase-2, oxidized LDL-induced activation required LOX-1, reactive oxygen species generation by NADPH oxidase and p38-MAPK activation. Inhibition of sphingosine kinase-1 blocked the angiogenic response and triggered HMEC-1 apoptosis. Low concentration of oxidized LDL was angiogenic in vivo, both in the Matrigel plug assay in mice and in the chorioallantoic membrane model, and was blocked by GW4869. In conclusion, low oxLDL concentration triggers sprouting angiogenesis that involves ROS-induced activation of the neutral sphingomyelinase-2/sphingosine kinase-1 pathway, and is effectively inhibited by GW4869.
Collapse
Affiliation(s)
- Caroline Camaré
- Inserm UMR-1048, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France; University of Toulouse, Faculty of Medicine, Biochemistry Department, Toulouse, France; CHU Toulouse, Rangueil, Toulouse, France
| | - Nathalie Augé
- Inserm UMR-1048, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | - Mélanie Pucelle
- Inserm UMR-1048, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | - Bertrand Saint-Lebes
- Inserm UMR-1048, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France; University of Toulouse, Faculty of Medicine, Biochemistry Department, Toulouse, France; CHU Toulouse, Rangueil, Toulouse, France
| | - Marie-Hélène Grazide
- University of Toulouse, Faculty of Medicine, Biochemistry Department, Toulouse, France
| | | | - Robert Salvayre
- Inserm UMR-1048, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France; University of Toulouse, Faculty of Medicine, Biochemistry Department, Toulouse, France; CHU Toulouse, Rangueil, Toulouse, France.
| |
Collapse
|
34
|
Qin W, Xie W, Xia N, He Q, Sun T. Silencing of Transient Receptor Potential Channel 4 Alleviates oxLDL-induced Angiogenesis in Human Coronary Artery Endothelial Cells by Inhibition of VEGF and NF-κB. Med Sci Monit 2016; 22:930-6. [PMID: 26999308 PMCID: PMC4805139 DOI: 10.12659/msm.897634] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Background Transient receptor potential channel 4 (TRPC4) plays central roles in endothelial cell function. The aim of this study was to investigate the silencing effects of TRPC4 on oxidized low-density lipoprotein (oxLDL)-induced angiogenesis in human coronary artery endothelial cells (HCAECs), as well as the underlying molecular mechanism involved in this process. Material/Methods HCAECs were transfected with small interfering RNA (siRNA) targeting TRPC4 (TRPC4-siRNA) or with a negative control (NC)-siRNA. The expression of TRPC4 was confirmed by real-time polymerase chain reaction (RT-PCR) and Western blotting. After the siRNA transfection, oxLDL was added to the medium. Cell proliferation, migration, and in vitro angiogenesis were determined by bromodeoxyuridine (BrdU) enzyme-linked immunosorbent assay (ELISA), Transwell assay and scratch-wound assay, respectively, and tube formation on Matrigel. Expression of vascular endothelial growth factor (VEGF) and nuclear factor (NF)-κB p65 were assessed by Western blotting. Results Both the mRNA and protein levels of TRPC4 were significantly reduced by transfection with TRPC4-siRNA compared to the control group or NC-siRNA group (P<0.05). Silencing of TRPC4 significantly decreased the cell proliferation, migration, and tube formation (all P<0.05). Furthermore, the expression levels of VEGF and NF-κB p65 were markedly lowered by silencing of TRPC4 in HCAECs. Conclusions These results suggest that silencing of TRPC4 alleviates angiogenesis induced by oxLDL in HCAECs through inactivation of VEGF and NF-κB. Suppression of TRPC4 might be an alternative therapeutic strategy for atherosclerotic neovascularization.
Collapse
Affiliation(s)
- Wen Qin
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Wei Xie
- Department of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Ning Xia
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Qinglin He
- , Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Tianwei Sun
- , Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
35
|
Dai Y, Zhang Z, Cao Y, Mehta JL, Li J. MiR-590-5p Inhibits Oxidized- LDL Induced Angiogenesis by Targeting LOX-1. Sci Rep 2016; 6:22607. [PMID: 26932825 PMCID: PMC4773867 DOI: 10.1038/srep22607] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/17/2016] [Indexed: 01/05/2023] Open
Abstract
Oxidized low-density lipoprotein (ox-LDL) is, at least in part, responsible for angiogenesis in atherosclerotic regions. This effect of ox-LDL has been shown to be mediated through a specific receptor LOX-1. Here we describe the effect of miR-590-5p on ox-LDL-mediated angiogenesis in in vitro and in vivo settings. Human umbilical vein endothelial cells (HUVECs) were transfected with miR-590-5p mimic or inhibitor followed by treatment with ox-LDL. In other experiments, Marigel plugs were inserted in the mice subcutaneous space. Both in vitro and in vivo studies showed that miR-590-5p mimic (100 nM) inhibited the ox-LDL-mediated angiogenesis (capillary tube formation, cell proliferation and migration as well as pro-angiogenic signals- ROS, MAPKs, pro-inflammatory cytokines and adhesion-related proteins). Of note, miR-590-5p inhibitor (200 nM) had the opposite effects. The inhibitory effect of miR-590-5p on angiogenesis was mediated by inhibition of LOX-1 at translational level. The inhibition of LOX-1 by miR-590-5p was confirmed by luciferase assay. In conclusion, we show that MiR-590-5p inhibits angiogenesis by targeting LOX-1 and suppressing redox-sensitive signals.
Collapse
Affiliation(s)
- Yao Dai
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, 230032 People's Republic of China.,Department of Medicine, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR 72205.,Department of Internal Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022 People's Republic of China
| | - Zhigao Zhang
- Department of Internal Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022 People's Republic of China
| | - Yongxiang Cao
- Department of Internal Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022 People's Republic of China
| | - Jawahar L Mehta
- Department of Medicine, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, 230032 People's Republic of China
| |
Collapse
|
36
|
Tsai CH, Yang MH, Hung AC, Wu SC, Chiu WC, Hou MF, Tyan YC, Wang YM, Yuan SSF. Identification of Id1 as a downstream effector for arsenic-promoted angiogenesis via PI3K/Akt, NF-κB and NOS signaling. Toxicol Res (Camb) 2015; 5:151-159. [PMID: 30090333 DOI: 10.1039/c5tx00280j] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 09/25/2015] [Indexed: 12/15/2022] Open
Abstract
Exposure to arsenic is known to be a risk factor for various types of cancer. Apart from its carcinogenic activity, arsenic also shows promoting effects on angiogenesis, a crucial process for tumor growth. Yet, the mechanism underlying arsenic-induced angiogenesis is not fully understood. In this study, we aimed at investigating the involvement of inhibitor of DNA binding 1 (Id1) and the associated signal molecules in the arsenic-mediated angiogenesis. Our initial screening revealed that treatment with low concentrations of arsenic (0.5-1 μM) led to multiple cellular responses, including enhanced endothelial cell viability and angiogenic activity as well as increased protein expression of Id1. The arsenic-induced angiogenesis was suppressed in the Id1-knocked down cells compared to that in control cells. Furthermore, arsenic-induced Id1 expression and angiogenic activity were regulated by PI3K/Akt, NF-κB, and nitric oxide synthase (NOS) signaling. In summary, our current data demonstrate for the first time that Id1 mediates the arsenic-promoted angiogenesis, and Id1 may be regarded as an antiangiogenesis target for treatment of arsenic-associated cancer.
Collapse
Affiliation(s)
- Chun-Hao Tsai
- Translational Research Center , Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan.,Graduate Institute of Medicine , College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan . ; Tel: +886-7-3121101 Ext2557
| | - Ming-Hui Yang
- Translational Research Center , Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan.,Department of Medical Research , Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Amos C Hung
- Translational Research Center , Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Shou-Cheng Wu
- Department of Biological Science and Technology and Institute of Molecular Medicine and Bioengineering , National Chiao Tung University , Hsinchu , Taiwan . ; Tel: +886-3-5712121 Ext56972
| | - Wen-Chin Chiu
- Division of Thoracic Surgery , Department of Surgery , Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Ming-Feng Hou
- Cancer Center , Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan.,Department of Surgery , Kaohsiung Municipal Ta-Tung Hospital , Kaohsiung , Taiwan
| | - Yu-Chang Tyan
- Translational Research Center , Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan.,Department of Medical Imaging and Radiological Sciences , Kaohsiung Medical University , Kaohsiung , Taiwan.,Center for Infectious Disease and Cancer Research , Kaohsiung Medical University , Kaohsiung , Taiwan.,Institute of Medical Science and Technology , National Sun Yat-sen University , Kaohsiung , Taiwan
| | - Yun-Ming Wang
- Department of Biological Science and Technology and Institute of Molecular Medicine and Bioengineering , National Chiao Tung University , Hsinchu , Taiwan . ; Tel: +886-3-5712121 Ext56972
| | - Shyng-Shiou F Yuan
- Translational Research Center , Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan.,Graduate Institute of Medicine , College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan . ; Tel: +886-7-3121101 Ext2557.,Department of Medical Research , Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan.,Department of Obstetrics and Gynecology , Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan.,Faculty and College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan.,Center for Lipid and Glycomedicine Research , Kaohsiung Medical University , Kaohsiung , Taiwan
| |
Collapse
|
37
|
Wang L, Li G, Chen Q, Ke D. Octanoylated ghrelin attenuates angiogenesis induced by oxLDL in human coronary artery endothelial cells via the GHSR1a-mediated NF-κB pathway. Metabolism 2015; 64:1262-71. [PMID: 26277200 DOI: 10.1016/j.metabol.2015.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 06/24/2015] [Accepted: 07/13/2015] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Low concentrations of oxidized low-density lipoprotein (oxLDL) promote the in vitro angiogenesis of endothelial cells and play an important role in plaque angiogenesis, which may cause plaque vulnerability and enhance the risk of intravascular thrombosis. The aim of this research was to investigate the effects of octanoylated ghrelin on oxLDL-induced angiogenesis and the underlying molecular mechanisms involved in this process. MATERIALS/METHODS Human coronary artery endothelial cells (HCAECs) were incubated with 5 μg/ml oxLDL and treated with various concentrations of octanoylated ghrelin (10(-9)-10(-6)M) with or without inhibitors for 24h. Cell proliferation, migration, and in vitro angiogenesis were analyzed by bromodeoxyuridine (BrdU) staining and BrdU enzyme-linked immunosorbent assay (ELISA), transwell assay, and tube formation on Matrigel, respectively. NF-κB (nuclear factor κB) expression was determined by Western-blot analysis. RESULTS Treatment with oxLDL at 5 μg/ml enhanced the proliferation, migration and tube formation of HCAECs. In contrast, pretreatment with octanoylated ghrelin significantly attenuated in vitro angiogenesis in oxLDL-induced HCAECs. In addition, Western blot analysis indicated that NF-κB expression was increased after oxLDL treatment, and that this effect was significantly reversed by pretreatment with octanoylated ghrelin. However, the NF-κB inhibitor PDTC or the GHSR1a inhibitor [D-Lys3]-GHRP-6 abolished the effects of octanoylated ghrelin on the inhibition of angiogenesis and NF-κB p65 expression induced by oxLDL. CONCLUSIONS These findings suggest that octanoylated ghrelin attenuates angiogenesis induced by oxLDL in HCAECs via the inhibition of GHSR1a-mediated NF-κB pathway. Furthermore, octanoylated ghrelin may promote the stability of vulnerable plaques by inhibiting plaque angiogenesis.
Collapse
Affiliation(s)
- Li Wang
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Guiqiong Li
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qingwei Chen
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Dazhi Ke
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
38
|
Li S, Li Q, Lv X, Liao L, Yang W, Li S, Lu P, Zhu D. Aurantio-obtusin relaxes systemic arteries through endothelial PI3K/AKT/eNOS-dependent signaling pathway in rats. J Pharmacol Sci 2015; 128:108-15. [PMID: 26076958 DOI: 10.1016/j.jphs.2015.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 05/15/2015] [Accepted: 05/19/2015] [Indexed: 02/05/2023] Open
Abstract
Aurantio-obtusin is a natural effective compound isolated from Semen Cassiae, which possesses hypotensive and hypolipidemic effects. Although its hypotensive effect have been clarified, mechanisms Aurantio-obtusin relaxes systemic arteries remain unclear. This study was to investigate effects and mechanisms of Aurantio-obtusin on isolated mesenteric arteries (MAs). We examined MAs relaxation induced by Aurantio-obtusin on rat isolated MAs, expression and activity of endothelial nitric oxide synthase (eNOS) and protein kinase B (AKT), and nitric oxide (NO) production in bovine artery endothelial cells (BAECs). Findings showed Aurantio-obtusin elicited dose-dependent vasorelaxation with phenylephrine (PE) precontracted rat MA rings (diameter: 200-300 μm), which can be diminished by denudation of endothelium and inhibition of eNOS activity, while having no effect on rat isolated pulmonary artery (PA) rings. Aurantio-obtusin increased NO production by promoting phosphorylations of eNOS at Ser-1177 and Thr-495 in endothelial cells. Aurantio-obtusin also promoted phosphorylations of Akt at Ser-473. PI3K inhibitor LY290042 could diminish vasorelaxation induced by Aurantio-obtusin. Moreover Aurantio-obtusin also elicited dose-dependent vasorelaxation effect with PE precontracted MA rings (diameter: 100-150 μm). Therefore, vasorelaxation induced by Aurantio-obtusin was dependent on endothelium integrity and NO production, which mediated by endothelial PI3K/Akt/eNOS pathway. Results suggest Aurantio-obtusin may offer therapeutic effects in hypertension, as a new potential vasodilator.
Collapse
Affiliation(s)
- Shuzhen Li
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, PR China
| | - Qian Li
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, PR China
| | - Xinyu Lv
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, PR China
| | - Lin Liao
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, PR China
| | - Weiwei Yang
- College of Food, Northeast Agriculture University, PR China
| | - Shanshan Li
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, PR China
| | - Ping Lu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, PR China
| | - Daling Zhu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, PR China.
| |
Collapse
|
39
|
Camaré C, Trayssac M, Garmy-Susini B, Mucher E, Sabbadini R, Salvayre R, Negre-Salvayre A. Oxidized LDL-induced angiogenesis involves sphingosine 1-phosphate: prevention by anti-S1P antibody. Br J Pharmacol 2014; 172:106-18. [PMID: 25176316 DOI: 10.1111/bph.12897] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 08/13/2014] [Accepted: 08/24/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Neovascularization occurring in atherosclerotic lesions may promote plaque expansion, intraplaque haemorrhage and rupture. Oxidized LDL (oxLDL) are atherogenic, but their angiogenic effect is controversial; both angiogenic and anti-angiogenic effects have been reported. The angiogenic mechanism of oxLDL is partly understood, but the role of the angiogenic sphingolipid, sphingosine 1-phosphate (S1P), in this process is not known. Thus, we investigated whether S1P is involved in the oxLDL-induced angiogenesis and whether an anti-S1P monoclonal antibody can prevent this effect. EXPERIMENTAL APPROACH Angiogenesis was assessed by capillary tube formation by human microvascular endothelial cells (HMEC-1) cultured on Matrigel and in vivo by the Matrigel plug assay in C57BL/6 mice. KEY RESULTS Human oxLDL exhibited a biphasic angiogenic effect on HMEC-1; low concentrations were angiogenic, higher concentrations were cytotoxic. The angiogenic response to oxLDL was blocked by the sphingosine kinase (SPHK) inhibitor, dimethylsphingosine, by SPHK1-siRNA and by an anti-S1P monoclonal antibody. Moreover, inhibition of oxLDL uptake and subsequent redox signalling by anti-CD36 and anti-LOX-1 receptor antibodies and by N-acetylcysteine, respectively, blocked SPHK1 activation and tube formation. In vivo, in the Matrigel plug assay, low concentrations of human oxLDL or murine oxVLDL also triggered angiogenesis, which was prevented by i.p. injection of the anti-S1P antibody. CONCLUSION AND IMPLICATIONS These data highlight the role of S1P in angiogenesis induced by oxLDL both in HMEC-1 cultured on Matrigel and in vivo in the Matrigel plug model in mice, and demonstrate that the anti-S1P antibody effectively blocks the angiogenic effect of oxLDL.
Collapse
Affiliation(s)
- Caroline Camaré
- Inserm UMR-1048, Toulouse, France; Department of Biochemistry, University of Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
40
|
Myeloperoxidase oxidized LDL interferes with endothelial cell motility through miR-22 and heme oxygenase 1 induction: possible involvement in reendothelialization of vascular injuries. Mediators Inflamm 2014; 2014:134635. [PMID: 25530680 PMCID: PMC4233670 DOI: 10.1155/2014/134635] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 09/09/2014] [Indexed: 01/19/2023] Open
Abstract
Cardiovascular disease linked to atherosclerosis is the leading cause of death worldwide. Atherosclerosis is mainly linked to dysfunction in vascular endothelial cells and subendothelial accumulation of oxidized forms of LDL. In the present study, we investigated the role of myeloperoxidase oxidized LDL (Mox-LDL) in endothelial cell dysfunction. We studied the effect of proinflammatory Mox-LDL treatment on endothelial cell motility, a parameter essential for normal vascular processes such as angiogenesis and blood vessel repair. This is particularly important in the context of an atheroma plaque, where vascular wall integrity is affected and interference with its repair could contribute to progression of the disease. We investigated in vitro the effect of Mox-LDL on endothelial cells angiogenic properties and we also studied the signalling pathways that could be affected by analysing Mox-LDL effect on the expression of angiogenesis-related genes. We report that Mox-LDL inhibits endothelial cell motility and tubulogenesis through an increase in miR-22 and heme oxygenase 1 expression. Our in vitro data indicate that Mox-LDL interferes with parameters associated with angiogenesis. They suggest that high LDL levels in patients would impair their endothelial cell capacity to cope with a damaged endothelium contributing negatively to the progression of the atheroma plaque.
Collapse
|
41
|
Xiao M, Men LN, Xu MG, Wang GB, Lv HT, Liu C. Berberine protects endothelial progenitor cell from damage of TNF-α via the PI3K/AKT/eNOS signaling pathway. Eur J Pharmacol 2014; 743:11-6. [DOI: 10.1016/j.ejphar.2014.09.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 09/15/2014] [Accepted: 09/16/2014] [Indexed: 01/09/2023]
|
42
|
ZHOU TIAN, ZHENG YIMING, QIU JUHUI, HU JIANJUN, SUN DAMING, TANG CHAOJUN, WANG GUIXUE. ENDOTHELIAL MECHANOTRANSDUCTION MECHANISMS FOR VASCULAR PHYSIOLOGY AND ATHEROSCLEROSIS. J MECH MED BIOL 2014. [DOI: 10.1142/s0219519414300063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Vascular physiology and disease progression, such as atherosclerosis, are mediated by hemodynamic force generated from blood flow. The hemodynamic force exerts on vascular endothelial cells (ECs), which could perceive the mechanical signals and transmit them into cell interior by multiple potential shear sensors, collectively known as mechanotransduction. However, we do not understand completely how these shear-sensitive components orchestrate physiological and atherosclerotic responses to shear stress. In this review, we provide an overview of biomechanical mechanisms underlying vascular physiology and atherosclerotic progression. Additionally, we summarize current evidences to illustrate that atherosclerotic lesions preferentially develop in arterial regions experiencing disturbance in blood flow, during which endothelial dysfunction is the initial event of atherosclerosis, inflammation plays dominant roles in atherosclerotic progression, and angiogenesis emerges as compensatory explanation for atherosclerotic plaque rupture. Especially in the presence of systemic risk factors (e.g., hyperlipidaemia, hypertension and hyperglycemia), the synergy between these systemic risk factors with hemodynamic factors aggravates atherosclerosis by co-stimulating some of these biomechanical events. Given the hemodynamic environment of vasculature, understanding how the rapid shear-mediated signaling, particularly in combination with systemic risk factors, contribute to atherosclerotic progression through endothelial dysfunction, inflammation and angiogenesis helps to elucidate the role for atherogenic shear stress in specifically localizing atherosclerotic lesions in arterial regions with disturbed flow.
Collapse
Affiliation(s)
- TIAN ZHOU
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - YIMING ZHENG
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - JUHUI QIU
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - JIANJUN HU
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - DAMING SUN
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - CHAOJUN TANG
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - GUIXUE WANG
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
43
|
Valls RM, Farràs M, Suárez M, Fernández-Castillejo S, Fitó M, Konstantinidou V, Fuentes F, López-Miranda J, Giralt M, Covas MI, Motilva MJ, Solà R. Effects of functional olive oil enriched with its own phenolic compounds on endothelial function in hypertensive patients. A randomised controlled trial. Food Chem 2014; 167:30-5. [PMID: 25148955 DOI: 10.1016/j.foodchem.2014.06.107] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 03/13/2014] [Accepted: 06/24/2014] [Indexed: 10/25/2022]
Abstract
UNLABELLED The additional health-promoting properties of functional virgin olive oil (FVOO) enriched with its own phenolic compounds (OOPC) versus the parental virgin olive oil (VOO) must be tested in appropriate human clinical trials. Our aim was to assess the effects of FVOO on endothelial function in hypertensive patients. Thirteen pre- and stage-1 hypertensive patients received a single dose of 30 mL of FVOO (OOPC=961 mg/kg) or VOO (OOPC=289 mg/kg) in a postprandial randomised, double blind, crossover trial. Endothelial function, measured as ischemic reactive hyperemia (IRH) and related biomarkers, were followed for 5h after consumption. Compared with VOO, FVOO increased IRH (P<0.05) and plasma Cmax of hydroxytyrosol sulphate, a metabolite of OOPC 2h postprandial (P=0.05). After FVOO ingestion, oxidised LDL decreased (P=0.010) in an inverse relationship with IRH AUC values (P=0.01). FVOO provided more benefits on endothelial function than a standard natural virgin olive oil in pre- and hypertensive patients. TRIAL REGISTRATION isrctn.org. Identifier ISRCTN03450153.
Collapse
Affiliation(s)
- Rosa-M Valls
- Research Unit on Lipids and Atherosclerosis, Servei de Medicina Interna, Hospital Universitari Sant Joan, IISPV, Universitat Rovira i Virgili, CIBER Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Marta Farràs
- Cardiovascular Risk and Nutrition Research Group, IMIM-Research Institut Hospital del Mar, CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Barcelona, Spain; PhD Program in Biochemistry, Molecular Biology and Biomedicine, Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Manuel Suárez
- Food Technology Department, XaRTA-UTPV, Escuela Técnica Superior de Ingeniería Agraria, Universitat de Lleida, Av/Alcalde Rovira Roure 191, 25198 Lleida, Spain
| | - Sara Fernández-Castillejo
- Research Unit on Lipids and Atherosclerosis, Servei de Medicina Interna, Hospital Universitari Sant Joan, IISPV, Universitat Rovira i Virgili, CIBER Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Montserrat Fitó
- Cardiovascular Risk and Nutrition Research Group, IMIM-Research Institut Hospital del Mar, CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Barcelona, Spain
| | - Valentini Konstantinidou
- Research Unit on Lipids and Atherosclerosis, Servei de Medicina Interna, Hospital Universitari Sant Joan, IISPV, Universitat Rovira i Virgili, CIBER Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Francisco Fuentes
- Lipid and Atherosclerosis Unit, IMIBIC, Reina Sofia University Hospital, University of Cordoba, CIBER Fisiopatologia Obesidad y Nutrición (CIBEROBN), Instituto Salud Carlos III, Córdoba, Spain
| | - José López-Miranda
- Lipid and Atherosclerosis Unit, IMIBIC, Reina Sofia University Hospital, University of Cordoba, CIBER Fisiopatologia Obesidad y Nutrición (CIBEROBN), Instituto Salud Carlos III, Córdoba, Spain
| | - Montserrat Giralt
- Research Unit on Lipids and Atherosclerosis, Servei de Medicina Interna, Hospital Universitari Sant Joan, IISPV, Universitat Rovira i Virgili, CIBER Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - María-Isabel Covas
- Cardiovascular Risk and Nutrition Research Group, IMIM-Research Institut Hospital del Mar, CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Barcelona, Spain
| | - María-José Motilva
- Food Technology Department, XaRTA-UTPV, Escuela Técnica Superior de Ingeniería Agraria, Universitat de Lleida, Av/Alcalde Rovira Roure 191, 25198 Lleida, Spain.
| | - Rosa Solà
- Research Unit on Lipids and Atherosclerosis, Servei de Medicina Interna, Hospital Universitari Sant Joan, IISPV, Universitat Rovira i Virgili, CIBER Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain.
| |
Collapse
|
44
|
Akt inhibitor MK2206 prevents influenza pH1N1 virus infection in vitro. Antimicrob Agents Chemother 2014; 58:3689-96. [PMID: 24752266 DOI: 10.1128/aac.02798-13] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The influenza pH1N1 virus caused a global flu pandemic in 2009 and continues manifestation as a seasonal virus. Better understanding of the virus-host cell interaction could result in development of better prevention and treatment options. Here we show that the Akt inhibitor MK2206 blocks influenza pH1N1 virus infection in vitro. In particular, at noncytotoxic concentrations, MK2206 alters Akt signaling and inhibits endocytic uptake of the virus. Interestingly, MK2206 is unable to inhibit H3N2, H7N9, and H5N1 viruses, indicating that pH1N1 evolved specific requirements for efficient infection. Thus, Akt signaling could be exploited further for development of better therapeutics against pH1N1 virus.
Collapse
|
45
|
Hutter R, Speidl WS, Valdiviezo C, Sauter B, Corti R, Fuster V, Badimon JJ. Macrophages transmit potent proangiogenic effects of oxLDL in vitro and in vivo involving HIF-1α activation: a novel aspect of angiogenesis in atherosclerosis. J Cardiovasc Transl Res 2013; 6:558-69. [PMID: 23661177 DOI: 10.1007/s12265-013-9469-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 04/25/2013] [Indexed: 02/01/2023]
Abstract
Neovascularization has been linked to the progression and vulnerability of atherosclerotic lesions. Angiogenesis is increased in lipid-rich plaque. Hypoxia-inducible factor alpha (HIF-1α) is a key transcriptional regulator responding to hypoxia and activating genes, which promote angiogenesis, among them vascular endothelial growth factor (VEGF). Oxidized low-density lipoprotein (oxLDL) is generated in lipid-rich plaque by oxidative stress. It triggers an inflammatory response and was traditionally thought to inhibit endothelial cells. New data, however, suggest that oxLDL can activate HIF-1α in monocytes in a hypoxia-independent fashion. We hypothesized that HIF-1α activation in monocyte-macrophages could transmit proangiogenic effects of oxLDL linking hyperlipidemia, inflammation, and angiogenesis in atherosclerosis. First, we examined the effect of oxLDL on HIF-1α and VEGF expression in monocyte-macrophages and on their proangiogenic effect on endothelial cells in vitro in a monocyte-macrophage/endothelial co-culture model. OxLDL strongly induced HIF-1α and VEGF in monocyte-macrophages and significantly increased tube formation in co-cultured endothelial cells. HIF-1α inhibition reversed this effect. Second, we demonstrated a direct proangiogenic effect of oxLDL in an in vivo angiogenesis assay. Again, HIF-1α inhibition abrogated the proangiogenic effect of oxLDL. Third, in a rabbit atherosclerosis model, we studied the effect of dietary lipid lowering on arterial HIF-1α and VEGF expression. The administration of low-lipid diet significantly reduced the expression of both HIF-1α and VEGF, resulting in decreased plaque neovascularization. Our data point to oxLDL as a proangiogenic agent linking hyperlipidemia, inflammation, and angiogenesis in atherosclerosis. This effect is dependent on macrophages and, at least in part, on the induction of the HIF-1α pathway.
Collapse
Affiliation(s)
- Randolph Hutter
- The Cardiovascular Institute, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Chen X, Chen Q, Wang L, Li G. Ghrelin induces cell migration through GHSR1a-mediated PI3K/Akt/eNOS/NO signaling pathway in endothelial progenitor cells. Metabolism 2013; 62:743-52. [PMID: 23218924 DOI: 10.1016/j.metabol.2012.09.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 09/16/2012] [Accepted: 09/24/2012] [Indexed: 01/18/2023]
Abstract
OBJECTIVE The purpose of this research was to investigate the effects of ghrelin on circulating endothelial progenitor cells (EPC) directional migration and its underlying molecular mechanisms involved in this process. MATERIALS/METHODS EPC were isolated from bone marrow of SD rats by using Percoll density gradient centrifugation, and characterized by double positive for acLDL-Dil uptake and FITC-UEA-1 binding and immunocytochemistry for CD34, CD133, vWF and Flk-1. EPC were treated with different concentrations of ghrelin (10(-9)~10(-6)M) with or without GHSR1a inhibitor [D-Lys3]-GHRP-6, PI3K inhibitor LY294002 and endothelial nitric oxide synthase (eNOS) inhibitor L-NAME, migration of EPC was detected by transwell assay, levels of phosphorylated and total Akt and eNOS were determined by Western-blot analysis and Nitric Oxide (NO) production was measured by Griess assay, respectively. RESULTS EPC were successfully obtained by Percoll density gradient centrifugation and ghrelin at 10(-8)M~10(-7)M promoted EPC migration. Ghrelin-induced EPC migration was accompanied by phosphorylation of Akt and eNOS, as well as an increase in NO production. These biochemical events and EPC directional migration induced by ghrelin were completely inhibited by GHSR-1a blocker [D-Lys3]-GHRP-6. PI3K inhibitor LY294002 attenuated ghrelin-induced EPC migration, phosphorylation of Akt and eNOS, and NO production. eNOS inhibitor L-NAME blocked ghrelin-induced EPC migration, phosphorylation of eNOS, and NO production, but had no effect on Akt phosphorylation. CONCLUSIONS These findings suggest that ghrelin stimulates EPC directional migration via GHSR1a-mediated PI3K/Akt/eNOS/NO signal pathway. It indicates that ghrelin may be used as a therapeutic strategy to treat ischemic diseases by promoting EPC directional migration.
Collapse
Affiliation(s)
- Xiaodong Chen
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | | | | | | |
Collapse
|
47
|
Na KH, Choi JH, Kim CH, Kim KS, Kim GJ. Altered expression of norepinephrine transporter and norepinephrine in human placenta cause pre-eclampsia through regulated trophoblast invasion. Clin Exp Reprod Med 2013; 40:12-22. [PMID: 23614111 PMCID: PMC3630288 DOI: 10.5653/cerm.2013.40.1.12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 03/05/2013] [Accepted: 03/14/2013] [Indexed: 11/18/2022] Open
Abstract
Objective We investigated the norepinephrine transporter (NET) expression in normal and pre-eclamptic placentas and analyzed the invasion activity of trophoblastic cells based on norepinephrine (NE)-NET regulation. Methods NET and NE expression levels were examined by western blot and enzyme-linked immunosorbent assay, respectively. Trophoblast invasion activity, depending on NE-NET regulation, was determined by NET-small interfering RNA (siRNA) and NET transfection into the human extravillous trophoblast cells with or without NE treatment and invasion rates were analyzed by zymography and an invasion assay. Results NET mRNA was expressed at a low level in pre-eclamptic placentas compared with normal placentas and NE concentration in maternal plasma increased significantly in pre-eclamptic women compared to normal pregnant women (p<0.05). NET gene upregulation and NE treatment stimulated trophoblast cell invasion up to 2.5-fold (p<0.05) by stimulating matrix metalloproteinase-9 activity via the phosphoinositol-3-kinase/AKT signaling pathway, whereas NET-siRNA with NE treatment reduced invasion rates. Conclusion NET expression is reduced by inadequate regulation of NE levels during placental development. This suggests that a complementary balance between NET and NE regulates trophoblast cell invasion activities during placental development.
Collapse
Affiliation(s)
- Kyu-Hwan Na
- Department of Biomedical Science, CHA University, Seoul, Korea
| | | | | | | | | |
Collapse
|
48
|
Fu R, Wang Q, Guo Q, Xu J, Wu X. XJP-1 protects endothelial cells from oxidized low-density lipoprotein-induced apoptosis by inhibiting NADPH oxidase subunit expression and modulating the PI3K/Akt/eNOS pathway. Vascul Pharmacol 2013; 58:78-86. [DOI: 10.1016/j.vph.2012.08.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 07/23/2012] [Accepted: 08/16/2012] [Indexed: 11/28/2022]
|
49
|
Oxidized LDL triggers pro-oncogenic signaling in human breast mammary epithelial cells partly via stimulation of MiR-21. PLoS One 2012; 7:e46973. [PMID: 23091604 PMCID: PMC3473036 DOI: 10.1371/journal.pone.0046973] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 09/10/2012] [Indexed: 11/28/2022] Open
Abstract
Dyslipidemia and obesity are primary risk factors for the development of atherosclerosis and are also epidemiologically linked to increased susceptibility to a variety of cancers including breast cancer. One of the prominent features of dyslipidemia is enhanced production of oxidized LDL (ox-LDL), which has been shown to be implicated in key steps of atherogenesis including inflammatory signaling and proliferation of vascular cells. In this study we analyzed the effects of ox-LDL in human mammary epithelial cells (MCF10A). MCF10A cells avidly internalized dil-ox-LDL and exhibited increased proliferative response to ox-LDL within the range of 1–50 µg/ml in a dose-dependent manner. Treatment of cells with 20 µg/ml ox-LDL for 2 and 12 hours was associated with upregulation of LOX-1 and CD36 scavenger receptors while MSR1 and CXLC16 receptors did not change. Ox-LDL-treated cells displayed significant upregulation of NADPH oxidases (subunits P22phox and P47phox), lipoxygenases-12 and -15, and cytoplasmic, but not mitochondrial, SOD. Ox-LDL also triggered phosphorylation of IκBα coupled with nuclear translocation of NF-κB and stimulated p44/42 MAPK, PI3K and Akt while intracellular PTEN (PI3K/Akt pathway inhibitor and target of miR-21) declined. Quantitative PCR revealed increased expression of hsa-miR-21 in ox-LDL treated cells coupled with inhibition of miR-21 target genes. Further, transfection of MCF10A cells with miR-21 inhibitor prevented ox-LDL mediated stimulation of PI3K and Akt. We conclude that, similarly to vascular cells, mammary epithelial cells respond to ox-LDL by upregulation of proliferative and pro-inflammatory signaling. We also report for the first time that part of ox-LDL triggered reactions in MCF10A cells is mediated by oncogenic hsa-miR-21 through inhibition of its target gene PTEN and consequent activation of PI3K/Akt pathway.
Collapse
|
50
|
OxLDL stimulates Id1 nucleocytoplasmic shuttling in endothelial cell angiogenesis via PI3K Pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1821:1361-9. [DOI: 10.1016/j.bbalip.2012.07.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 07/12/2012] [Accepted: 07/16/2012] [Indexed: 12/26/2022]
|