1
|
Chang J, Yao Y, Sun X, Wang W, Qian H, Liu Y, Xue C, Ye W, Jiang F. JAG1 mediates apoptosis in herpes simplex keratitis by suppressing autophagy via ROS/JAG1/NOTCH1/pULK1 signaling pathway. Cell Biol Toxicol 2024; 41:1. [PMID: 39704867 PMCID: PMC11662045 DOI: 10.1007/s10565-024-09968-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Herpes simplex keratitis (HSK), an ocular disease resulted from herpes simplex virus type 1 (HSV-1) infection, leads to the majority of infectious corneal blindness worldwide. The apoptosis of corneal epithelial cells (CECs) resulted from HSV-1 disrupts the epithelial barrier and exacerbates the infection; however, there is no definitive cure for HSK. Jagged1 (JAG1), one of the primary functional ligands for NOTCH receptors, plays a crucial role in regulating apoptosis and autophagy; however, its role in HSK is unclear. Our transcriptome analysis showed JAG1 was significantly upregulated in HSV-1-infected human CECs. We aimed to explore JAG1's role in regulating apoptosis in HSV-1-infected human CECs and in HSK mice. HSV-1 infection induced apoptosis and reactive oxygen species (ROS) generation in CECs. HSV-1 also activated the JAG1/NOTCH1 signaling pathway. The ROS scavenger N-acetylcysteine significantly mitigated these effects. Additionally, inhibiting the JAG1/NOTCH1 pathway with short hairpin RNA against JAG1 or a NOTCH1 inhibitor (N-[N-{3,5-difuorophenacetyl}-1-alanyl]-S-phenylglycine t-butyl ester [DAPT]) alleviated HSV-1-induced CEC apoptosis. Transmission electron microscopy and western blotting revealed that HSV-1 infection suppressed ULK1-mediated autophagy in CECs, while DAPT treatment enhanced autophagy by suppressing ULK1 phosphorylation. The activation of autophagy by rapamycin treatment markedly reduced ROS levels and apoptosis in HSV-1-infected CECs, revealing a synergistic effect between the suppressed autophagy and increased ROS levels, ultimately leading to apoptosis. Thus, HSV-1 induces CEC apoptosis by suppressing autophagy through ROS/JAG1/NOTCH1/pULK1 signaling pathway in vitro and in vivo, providing potential therapeutic targets for HSK.
Collapse
Affiliation(s)
- Jingyao Chang
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Yao Yao
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Xinghong Sun
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Nanjing, 210011, Jiangsu, China
| | - Wenzhe Wang
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Haochen Qian
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Yumeilan Liu
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Chunyan Xue
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
- Department of Ophthalmology, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, Hainan, China.
| | - Wei Ye
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China.
| | - Feng Jiang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Nanjing, 210011, Jiangsu, China.
| |
Collapse
|
2
|
Lian Z, Kuerban R, Niu Z, Aisaiti P, Wu C, Yang X. Notch Signaling Is Associated with Pulmonary Fibrosis in Patients with Pigeon Breeder's Lung by Regulating Oxidative Stress. Emerg Med Int 2024; 2024:7610032. [PMID: 39139588 PMCID: PMC11321885 DOI: 10.1155/2024/7610032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/03/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
This study explored the molecular mechanism underlying the association of Notch signaling and oxidative stress with the occurrence of pulmonary fibrosis in patients with pigeon breeder's lung (PBL). Rat models of fibrotic PBL were constructed with freeze-dried protein powder, and the animals were divided into the control (intratracheal instillation of normal saline; n = 9), M (PBL model; intratracheal instillation of freeze-dried protein powder; n = 9), and M + D (PBL+ the Notch inhibitor DAPT; n = 9) groups. Immunohistochemistry was employed to observe the protein levels of pathway factors and α-SMA, and the levels of ROS, GSH-PX, SOD, and MDA were observed using ELISA. To verify the results of the animal experiment, cytological models were constructed. The M group and the M + D group had significantly increased α-SMA levels (P < 0.05). Although both groups had significantly higher key protein levels in the Notch channel, the M + D group had significantly lower levels relative to the M group (P < 0.05). Oxidative stress products were examined, and the levels of MDA and ROS were significantly increased, while those of GSH-PX and SOD were significantly decreased in the M and M + D groups as compared to the control, but the M group and the M + D group significantly differed (P < 0.05). These findings were further validated by the cytological experiment. Notch signaling is associated with pulmonary fibrosis in PBL by regulating cellular oxidative stress, and inhibiting this pathway can slow down pulmonary fibrosis progression.
Collapse
Affiliation(s)
- Zhichuang Lian
- Graduate SchoolXinjiang Medical University, Urumqi 830001, China
| | - Remila Kuerban
- Department of Respiratory and Critical Care MedicinePeople's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, China
| | - Zongxin Niu
- Department of Respiratory and Critical Care MedicinePeople's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, China
| | - Paruzha Aisaiti
- Department of Respiratory and Critical Care MedicinePeople's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, China
| | - Chao Wu
- Graduate SchoolXinjiang Medical University, Urumqi 830001, China
| | - Xiaohong Yang
- Graduate SchoolXinjiang Medical University, Urumqi 830001, China
| |
Collapse
|
3
|
Wei M, Lu Z, Zhang H, Fan X, Zhang X, Jiang B, Li J, Xue M. Aspirin and Celecoxib Regulate Notch1/Hes1 Pathway to Prevent Pressure Overload-Induced Myocardial Hypertrophy. Int Heart J 2024; 65:475-486. [PMID: 38825493 DOI: 10.1536/ihj.23-614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
This study aimed to investigate the molecular mechanisms underlying the protective effects of cyclooxygenase (cox) inhibitors against myocardial hypertrophy.Rat H9c2 cardiomyocytes were induced by mechanical stretching. SD rats underwent transverse aortic constriction to induce pressure overload myocardial hypertrophy. Rats were subjected to echocardiography and tail arterial pressure in 12W. qPCR and western blot were used to detect the expression of Notch-related signaling. The inflammatory factors were tested by ELISA in serum, heart tissue, and cell culture supernatant.Compared with control, levels of pro-inflammatory cytokines IL-6, TNF-α, and IL-1β were increased and anti-inflammatory cytokine IL-10 was reduced in myocardial tissues and serum of rat models. Levels of Notch1 and Hes1 were reduced in myocardial tissues. However, cox inhibitor treatment (aspirin and celecoxib), the improvement of exacerbated myocardial hypertrophy, fibrosis, dysfunction, and inflammation was parallel to the activation of Notch1/Hes1 pathway. Moreover, in vitro experiments showed that, in cardiomyocyte H9c2 cells, application of ~20% mechanical stretching activated inflammatory mediators (IL-6, TNF-α, and IL-1β) and hypertrophic markers (ANP and BNP). Moreover, expression levels of Notch1 and Hes1 were decreased. These changes were effectively alleviated by aspirin and celecoxib.Cox inhibitors may protect heart from hypertrophy and inflammation possibly via the Notch1/Hes1 signaling pathway.
Collapse
Affiliation(s)
- Minghui Wei
- School of Basic Medicine, Inner Mongolia Medical University
| | - Ziyu Lu
- School of Basic Medicine, Inner Mongolia Medical University
| | - Haifeng Zhang
- Office of Academic Affairs, Inner Mongolia Medical University
| | - Xiaomei Fan
- Department of Physiology, Inner Mongolia Medical University
| | - Xin Zhang
- Department of Physiology, Inner Mongolia Medical University
| | - Bihui Jiang
- School of Basic Medicine, Inner Mongolia Medical University
| | - Jianying Li
- School of Basic Medicine, Inner Mongolia Medical University
| | - Mingming Xue
- Office of Academic Affairs, Inner Mongolia Medical University
| |
Collapse
|
4
|
Zhang YH, Wang T, Li YF, Deng YN, He XL, Wang LJ. N-acetylcysteine improves autism-like behavior by recovering autophagic deficiency and decreasing Notch-1/Hes-1 pathway activity. Exp Biol Med (Maywood) 2023; 248:966-978. [PMID: 37377100 PMCID: PMC10525405 DOI: 10.1177/15353702231179924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 04/16/2023] [Indexed: 06/29/2023] Open
Abstract
N-acetylcysteine (NAC) has been reported to improve social interaction behavior, irritability, self-injury, and anxiety-like behavior in autism. However, the molecular mechanism underlying the therapeutic roles of NAC in autism remains unknown. This study mainly aimed to investigate the therapeutic effect of NAC on valproic acid (VPA)-induced autism model and the underlying mechanisms. Our results showed that NAC ameliorated the deficits in sociability and the anxiety- and repetitive-like behaviors displayed by VPA-exposed rats. In addition, VPA exposure induced autophagic deficiency and enhanced Notch-1/Hes-1 pathway activity based on lowered Beclin-1 and LC3B levels, while increased expression of p62, Notch-1, and Hes-1 expression at the protein level. However, NAC recovered VPA-induced autophagic deficiency and reduced Notch-1/Hes-1 pathway activity in a VPA-exposed autism rat model and SH-SY5Y neural cells. The present results demonstrated that NAC improves autism-like behavioral abnormalities by inactivating Notch-1/Hes-1 signaling pathway and recovering autophagic deficiency. Taken together, this study helps to elucidate a novel molecular mechanism that underlies the therapeutic actions of NAC in autism and suggests its potential to ameliorate behavioral abnormalities in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ying-Hua Zhang
- Department of Human Anatomy & Histoembryology, Henan Key Laboratory of Biological Psychiatry, School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang 453003, China
| | - Ting Wang
- Department of Human Anatomy & Histoembryology, Henan Key Laboratory of Biological Psychiatry, School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang 453003, China
| | - Yan-Fang Li
- Department of Human Anatomy & Histoembryology, Henan Key Laboratory of Biological Psychiatry, School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang 453003, China
| | - Ya-Nan Deng
- Department of Human Anatomy & Histoembryology, Henan Key Laboratory of Biological Psychiatry, School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang 453003, China
| | - Xue-Ling He
- Department of Human Anatomy & Histoembryology, Henan Key Laboratory of Biological Psychiatry, School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang 453003, China
| | - Li-Jun Wang
- Department of Human Anatomy & Histoembryology, Henan Key Laboratory of Biological Psychiatry, School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
5
|
Kotlyarov S. Effects of Atherogenic Factors on Endothelial Cells: Bioinformatics Analysis of Differentially Expressed Genes and Signaling Pathways. Biomedicines 2023; 11:1216. [PMID: 37189834 PMCID: PMC10135807 DOI: 10.3390/biomedicines11041216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
(1) Background: Atherosclerosis is a serious medical condition associated with high morbidity and mortality rates. It develops over many years as a complex chain of events in the vascular wall involving various cells and is influenced by many factors of clinical interest. (2) Methods: In this study, we performed a bioinformatic analysis of Gene Expression Omnibus (GEO) datasets to investigate the gene ontology of differentially expressed genes (DEGs) in endothelial cells exposed to atherogenic factors such as tobacco smoking, oscillatory shear, and oxidized low-density lipoproteins (oxLDL). DEGs were identified using the limma R package, and gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, and protein-protein interaction (PPI) network analysis were performed. (3) Results: We studied biological processes and signaling pathways involving DEGs in endothelial cells under the influence of atherogenic factors. GO enrichment analysis demonstrated that the DEGs were mainly involved in cytokine-mediated signaling pathway, innate immune response, lipid biosynthetic process, 5-lipoxygenase activity, and nitric-oxide synthase activity. KEGG pathway enrichment analysis showed that common pathways included tumor necrosis factor signaling pathway, NF-κB signaling pathway, NOD-like receptor signaling pathway, lipid and atherosclerosis, lipoprotein particle binding, and apoptosis. (4) Conclusions: Atherogenic factors such as smoking, impaired flow, and oxLDL contribute to impaired innate immune response, metabolism, and apoptosis in endothelial cells, potentially leading to the development of atherosclerosis.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
6
|
Agri-Food Waste from Apple, Pear, and Sugar Beet as a Source of Protective Bioactive Molecules for Endothelial Dysfunction and Its Major Complications. Antioxidants (Basel) 2022; 11:antiox11091786. [PMID: 36139860 PMCID: PMC9495678 DOI: 10.3390/antiox11091786] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Endothelial damage is recognized as the initial step that precedes several cardiovascular diseases (CVD), such as atherosclerosis, hypertension, and coronary artery disease. It has been demonstrated that the best treatment for CVD is prevention, and, in the frame of a healthy lifestyle, the consumption of vegetables, rich in bioactive molecules, appears effective at reducing the risk of CVD. In this context, the large amount of agri-food industry waste, considered a global problem due to its environmental and economic impact, represents an unexplored source of bioactive compounds. This review provides a summary regarding the possible exploitation of waste or by-products derived by the processing of three traditional Italian crops-apple, pear, and sugar beet-as a source of bioactive molecules to protect endothelial function. Particular attention has been given to the bioactive chemical profile of these pomaces and their efficacy in various pathological conditions related to endothelial dysfunction. The waste matrices of apple, pear, and sugar beet crops can represent promising starting material for producing "upcycled" products with functional applications, such as the prevention of endothelial dysfunction linked to cardiovascular diseases.
Collapse
|
7
|
Liu J, Huang L, Wan M, Chen G, Su M, Han F, Liu F, Xiong G, Liao X, Lu H, Li W, Cao Z. Lenvatinib induces cardiac developmental toxicity in zebrafish embryos through regulation of Notch mediated-oxidative stress generation. ENVIRONMENTAL TOXICOLOGY 2022; 37:1310-1320. [PMID: 35119177 DOI: 10.1002/tox.23485] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/17/2022] [Accepted: 01/22/2022] [Indexed: 06/14/2023]
Abstract
Due to an increasing number of abused drugs dumped into the wastewater, more and more drugs are detected in the water environment, which may affect the survival of aquatic organisms. Lenvatinib is a multi-targeted tyrosine kinase inhibitor, and is clinically used to treat differentiated thyroid cancer, renal epithelial cell carcinoma and liver cancer. However, there are few reports on the effects of lenvatinib in embryos development. In this study, zebrafish embryos were used to evaluate the effect of lenvatinib on cardiovascular development. Well-developed zebrafish embryos were selected at 6 h post fertilization (hpf) and exposed to 0.05 mg/L, 0.1 mg/L and 0.2 mg/L lenvatinib up to 72 hpf. The processed embryos demonstrated cardiac edema, decreased heart rate, prolonged SV-BA distance, inhibited angiogenesis, and blocked blood circulation. Lenvatinib caused cardiac defects in the whole stage of cardiac development and increased the apoptosis of cardiomyocyte. Oxidative stress in the processed embryos was accumulated and inhibiting oxidative stress could rescue cardiac defects induced by lenvatinib. Additionally, we found that lenvatinib downregulated Notch signaling, and the activation of Notch signaling could rescue cardiac developmental defects and downregulate oxidative stress level induced by lenvatinib. Our results suggested that lenvatinib might induce cardiac developmental toxicity through inducing Notch mediated-oxidative stress generation, raising concerns about the harm of exposure to lenvatinib in aquatic organisms.
Collapse
Affiliation(s)
- Jieping Liu
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, Fujian, China
| | - Ling Huang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, Fujian, China
| | - Mengqi Wan
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jinggangshan University, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi, China
| | - Guilan Chen
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jinggangshan University, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi, China
| | - Meile Su
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jinggangshan University, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi, China
| | - Fang Han
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, Fujian, China
| | - Fasheng Liu
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jinggangshan University, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi, China
| | - Guanghua Xiong
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jinggangshan University, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi, China
| | - Xinjun Liao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jinggangshan University, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi, China
| | - Huiqiang Lu
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jinggangshan University, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi, China
| | - Wanbo Li
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, Fujian, China
| | - Zigang Cao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jinggangshan University, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi, China
| |
Collapse
|
8
|
Chung J, Kim KH, Yu N, An SH, Lee S, Kwon K. Fluid Shear Stress Regulates the Landscape of microRNAs in Endothelial Cell-Derived Small Extracellular Vesicles and Modulates the Function of Endothelial Cells. Int J Mol Sci 2022; 23:ijms23031314. [PMID: 35163238 PMCID: PMC8836123 DOI: 10.3390/ijms23031314] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 12/10/2022] Open
Abstract
Blood fluid shear stress (FSS) modulates endothelial function and vascular pathophysiology. The small extracellular vesicles (sEVs) such as exosomes are potent mediators of intercellular communication, and their contents reflect cellular stress. Here, we explored the miRNA profiles in endothelial cells (EC)-derived sEVs (EC-sEVs) under atheroprotective laminar shear stress (LSS) and atheroprone low-oscillatory shear stress (OSS) and conducted a network analysis to identify the main biological processes modulated by sEVs’ miRNAs. The EC-sEVs were collected from culture media of human umbilical vein endothelial cells exposed to atheroprotective LSS (20 dyne/cm2) and atheroprone OSS (±5 dyne/cm2). We explored the miRNA profiles in FSS-induced EC-sEVs (LSS-sEVs and OSS-sEVs) and conducted a network analysis to identify the main biological processes modulated by sEVs’ miRNAs. In vivo studies were performed in a mouse model of partial carotid ligation. The sEVs’ miRNAs-targeted genes were enriched for endothelial activation such as angiogenesis, cell migration, and vascular inflammation. OSS-sEVs promoted tube formation, cell migration, monocyte adhesion, and apoptosis, and upregulated the expression of proteins that stimulate these biological processes. FSS-induced EC-sEVs had the same effects on endothelial mechanotransduction signaling as direct stimulation by FSS. In vivo studies showed that LSS-sEVs reduced the expression of pro-inflammatory genes, whereas OSS-sEVs had the opposite effect. Understanding the landscape of EC-exosomal miRNAs regulated by differential FSS patterns, this research establishes their biological functions on a system level and provides a platform for modulating the overall phenotypic effects of sEVs.
Collapse
Affiliation(s)
- Jihwa Chung
- Exollence Biotechnology Co., Ltd., Seoul 07985, Korea; (J.C.); (K.H.K.); (S.H.A.)
| | - Kyoung Hwa Kim
- Exollence Biotechnology Co., Ltd., Seoul 07985, Korea; (J.C.); (K.H.K.); (S.H.A.)
| | - Namhee Yu
- Research Institute, National Cancer Center, Goyangsi 10408, Korea;
| | - Shung Hyun An
- Exollence Biotechnology Co., Ltd., Seoul 07985, Korea; (J.C.); (K.H.K.); (S.H.A.)
| | - Sanghyuk Lee
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea;
| | - Kihwan Kwon
- Exollence Biotechnology Co., Ltd., Seoul 07985, Korea; (J.C.); (K.H.K.); (S.H.A.)
- Department of Internal Medicine, Cardiology Division, School of Medicine, Ewha Womans University, Seoul 07985, Korea
- Correspondence: ; Tel.: +82-2-2650-2640
| |
Collapse
|
9
|
Qin X, Zhang K, Qiu J, Wang N, Qu K, Cui Y, Huang J, Luo L, Zhong Y, Tian T, Wu W, Wang Y, Wang G. Uptake of oxidative stress-mediated extracellular vesicles by vascular endothelial cells under low magnitude shear stress. Bioact Mater 2021; 9:397-410. [PMID: 34820579 PMCID: PMC8586717 DOI: 10.1016/j.bioactmat.2021.10.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/11/2021] [Accepted: 10/20/2021] [Indexed: 11/17/2022] Open
Abstract
Extracellular vesicles (EVs) are increasingly used as delivery vehicles for drugs and bioactive molecules, which usually require intravascular administration. The endothelial cells covering the inner surface of blood vessels are susceptible to the shear stress of blood flow. Few studies demonstrate the interplay of red blood cell-derived EVs (RBCEVs) and endothelial cells. Thus, the phagocytosis of EVs by vascular endothelial cells during blood flow needs to be elucidated. In this study, red blood cell-derived extracellular vesicles (RBCEVs) were constructed to investigate endothelial cell phagocytosis in vitro and animal models. Results showed that low magnitude shear stress including low shear stress (LSS) and oscillatory shear stress (OSS) could promote the uptake of RBCEVs by endothelial cells in vitro. In addition, in zebrafish and mouse models, RBCEVs tend to be internalized by endothelial cells under LSS or OSS. Moreover, RBCEVs are easily engulfed by endothelial cells in atherosclerotic plaques exposed to LSS or OSS. In terms of mechanism, oxidative stress induced by LSS is part of the reason for the increased uptake of endothelial cells. Overall, this study shows that vascular endothelial cells can easily engulf EVs in areas of low magnitude shear stress, which will provide a theoretical basis for the development and utilization of EVs-based nano-drug delivery systems in vivo. We recently reported that endothelial cells were amateur phagocytic cells for RBCEVs engulfment. Low magnitude shear stress (LSS and OSS) can increase the uptake of RBCEVs by endothelial cells in vitro and in vivo. ROS induced by low magnitude shear stress acts as an accelerator to enhance endothelial cells uptake of RBCEVs.
Collapse
Affiliation(s)
- Xian Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Nan Wang
- The Nanoscience Centre, University of Cambridge, Cambridge, CB3 0FF, UK
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yuliang Cui
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Junli Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Li Luo
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yuan Zhong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Tian Tian
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| |
Collapse
|
10
|
Fortini F, Vieceli Dalla Sega F, Marracino L, Severi P, Rapezzi C, Rizzo P, Ferrari R. Well-Known and Novel Players in Endothelial Dysfunction: Updates on a Notch(ed) Landscape. Biomedicines 2021; 9:biomedicines9080997. [PMID: 34440201 PMCID: PMC8393382 DOI: 10.3390/biomedicines9080997] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 12/27/2022] Open
Abstract
Endothelial dysfunction characterizes every aspect of the so-called cardiovascular continuum, a series of events ranging from hypertension to the development of atherosclerosis and, finally, to coronary heart disease, thrombus formation, myocardial infarction, and heart failure. Endothelial dysfunction is the main prognostic factor for the progression of vascular disorders, which responds to drug intervention and lifestyle changes. Virtually all of the drugs used to prevent cardiovascular disorders, such as long-used and new antilipidemic agents and inhibitors of angiotensin enzyme (ACEi), exert an important effect on the endothelium. Endothelial dysfunction is a central feature of coronavirus disease -19 (COVID-19), and it is now clear that life-risk complications of the disease are prompted by alterations of the endothelium induced by viral infection. As a consequence, the progression of COVID-19 is worse in the subjects in whom endothelial dysfunction is already present, such as elderly, diabetic, obese, and hypertensive patients. Importantly, circulating biomarkers of endothelial activation and injury predict the severity and mortality of the disease and can be used to evaluate the efficacy of treatments. The purpose of this review is to provide updates on endothelial function by discussing its clinical relevance in the cardiovascular continuum, the latest insights from molecular and cellular biology, and their implications for clinical practice, with a focus on new actors, such as the Notch signaling and emerging therapies for cardiovascular disease.
Collapse
Affiliation(s)
- Francesca Fortini
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
| | | | - Luisa Marracino
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Paolo Severi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Claudio Rapezzi
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Roberto Ferrari
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
- Correspondence: ; Tel.: +39-053-229-3707
| |
Collapse
|
11
|
COVID-19: Direct and Indirect Mechanisms of Statins. Int J Mol Sci 2021; 22:ijms22084177. [PMID: 33920709 PMCID: PMC8073792 DOI: 10.3390/ijms22084177] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/10/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023] Open
Abstract
The virus responsible for the current COVID-19 pandemic is severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2): a new virus with high infectivity and moderate mortality. The major clinical manifestation of COVID-19 is interstitial pneumonia, which may progress to acute respiratory distress syndrome (ARDS). However, the disease causes a potent systemic hyperin-flammatory response, i.e., a cytokine storm or macrophage activation syndrome (MAS), which is associated with thrombotic complications. The complexity of the disease requires appropriate intensive treatment. One of promising treatment is statin administration, these being 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors that exert pleiotropic anti-inflammatory effects. Recent studies indicate that statin therapy is associated with decreased mortality in COVID-19, which may be caused by direct and indirect mechanisms. According to literature data, statins can limit SARS-CoV-2 cell entry and replication by inhibiting the main protease (Mpro) and RNA-dependent RNA polymerase (RdRp). The cytokine storm can be ameliorated by lowering serum IL-6 levels; this can be achieved by inhibiting Toll-like receptor 4 (TLR4) and modulating macrophage activity. Statins can also reduce the complications of COVID-19, such as thrombosis and pulmonary fibrosis, by reducing serum PAI-1 levels, attenuating TGF-β and VEGF in lung tissue, and improving endothelial function. Despite these benefits, statin therapy may have side effects that should be considered, such as elevated creatinine kinase (CK), liver enzyme and serum glucose levels, which are already elevated in severe COVID-19 infection. The present study analyzes the latest findings regarding the benefits and limitations of statin therapy in patients with COVID-19.
Collapse
|
12
|
Li J, Han R, Li J, Zhai L, Xie X, Zhang J, Chen Y, Luo J, Wang S, Sun Z, Cao Y, Lin L, Yang Q. Analysis of Molecular Mechanism of YiqiChutan Formula Regulating DLL4-Notch Signaling to Inhibit Angiogenesis in Lung Cancer. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8875503. [PMID: 33628824 PMCID: PMC7895574 DOI: 10.1155/2021/8875503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/11/2021] [Accepted: 01/30/2021] [Indexed: 12/26/2022]
Abstract
In order to explore the specific mechanism of YiqiChutan formula (YQCTF) in inhibiting the angiogenesis of lung cancer and its relationship with delta-like ligand 4- (DLL4-) Notch signaling, 30 healthy BALB/c-nu/nu rats were selected and divided into three groups: A549 group (implanted with lung adenocarcinoma cell line A549), NCI-H460 group (implanted with human lung large-cell carcinoma cell line NCI-H460), and NCI-H446 group (implanted with human lung small cell carcinoma cell line NCI-H446) for constructing lung cancer transplanted tumor models. After modeling, the group treated with normal saline was taken as control group, 200 mg/kg of YQCTF was adopted for intervention, and the tumor volume and growth inhibition rate were compared with the vascular targeted inhibitor Sorafenib. HE staining, CD31 fluorescent antibody staining, and microelectron microscopy were adopted to observe the neovascular endothelial cells of the transplanted tumor. The expression of VEGF, HIF-1α, DLL4, and Notch-1 in the transplanted tumors in each group was detected by Western blot and RT-PCR at the protein level or mRNA level. Compared with the control group, the YQCTF-treated group had obvious inhibitory effect on lung cancer transplanted tumor and lung cancer angiogenesis. In the YQCTF-treated group, the density of angiogenesis decreased significantly and the vascular lumen structure also decreased, and the expression levels of VEGF, HIF-1α, DLL4, and Notch-1 in the YQCTF-treated group were all lower than those in the control group. YQCTF could inhibit the growth of lung cancer transplanted tumor through antiangiogenesis, and it could also reduce the amount of angiogenesis in lung cancer transplanted tumor. In addition, the generation of lumen structure was also hindered, which was realized through the VEGF signaling pathway and DLL4-Notch signaling pathway.
Collapse
Affiliation(s)
- Jiayin Li
- Cancer Center, The First Affiliated Hospital to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Rui Han
- Cancer Center, The First Affiliated Hospital to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jing Li
- Department of Oncology, The First Affiliated Hospital to Hunan University of Chinese Medicine, Changsha 410000, China
| | - Linzhu Zhai
- Cancer Center, The First Affiliated Hospital to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xinying Xie
- Department of General, The First Affiliated Hospital to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jing Zhang
- Cancer Center, The First Affiliated Hospital to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yao Chen
- Cancer Center, The First Affiliated Hospital to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiamin Luo
- Cancer Center, The First Affiliated Hospital to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Sisi Wang
- Cancer Center, The First Affiliated Hospital to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhe Sun
- Cancer Center, The First Affiliated Hospital to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yang Cao
- Cancer Center, The First Affiliated Hospital to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Lizhu Lin
- The First Affiliated Hospital to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qiuye Yang
- Department of Medical Technologic, The First Affiliated Hospital to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
13
|
Vieceli Dalla Sega F, Fortini F, Cimaglia P, Marracino L, Tonet E, Antonucci A, Moscarelli M, Campo G, Rizzo P, Ferrari R. COX-2 Is Downregulated in Human Stenotic Aortic Valves and Its Inhibition Promotes Dystrophic Calcification. Int J Mol Sci 2020; 21:ijms21238917. [PMID: 33255450 PMCID: PMC7727817 DOI: 10.3390/ijms21238917] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/12/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the result of maladaptive fibrocalcific processes leading to a progressive thickening and stiffening of aortic valve (AV) leaflets. CAVD is the most common cause of aortic stenosis (AS). At present, there is no effective pharmacotherapy in reducing CAVD progression; when CAVD becomes symptomatic it can only be treated with valve replacement. Inflammation has a key role in AV pathological remodeling; hence, anti-inflammatory therapy has been proposed as a strategy to prevent CAVD. Cyclooxygenase 2 (COX-2) is a key mediator of the inflammation and it is the target of widely used anti-inflammatory drugs. COX-2-inhibitor celecoxib was initially shown to reduce AV calcification in a murine model. However, in contrast to these findings, a recent retrospective clinical analysis found an association between AS and celecoxib use. In the present study, we investigated whether variations in COX-2 expression levels in human AVs may be linked to CAVD. We extracted total RNA from surgically explanted AVs from patients without CAVD or with CAVD. We found that COX-2 mRNA was higher in non-calcific AVs compared to calcific AVs (0.013 ± 0.002 vs. 0.006 ± 0.0004; p < 0.0001). Moreover, we isolated human aortic valve interstitial cells (AVICs) from AVs and found that COX-2 expression is decreased in AVICs from calcific valves compared to AVICs from non-calcific AVs. Furthermore, we observed that COX-2 inhibition with celecoxib induces AVICs trans-differentiation towards a myofibroblast phenotype, and increases the levels of TGF-β-induced apoptosis, both processes able to promote the formation of calcific nodules. We conclude that reduced COX-2 expression is a characteristic of human AVICs prone to calcification and that COX-2 inhibition may promote aortic valve calcification. Our findings support the notion that celecoxib may facilitate CAVD progression.
Collapse
Affiliation(s)
| | - Francesca Fortini
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.V.D.S.); (F.F.); (P.C.); (M.M.); (R.F.)
| | - Paolo Cimaglia
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.V.D.S.); (F.F.); (P.C.); (M.M.); (R.F.)
| | - Luisa Marracino
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy;
| | - Elisabetta Tonet
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Cona, Italy; (E.T.); (A.A.); (G.C.)
| | - Antonio Antonucci
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Cona, Italy; (E.T.); (A.A.); (G.C.)
| | - Marco Moscarelli
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.V.D.S.); (F.F.); (P.C.); (M.M.); (R.F.)
| | - Gianluca Campo
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Cona, Italy; (E.T.); (A.A.); (G.C.)
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.V.D.S.); (F.F.); (P.C.); (M.M.); (R.F.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy;
- Correspondence: ; Tel.: +39-0532-455-508
| | - Roberto Ferrari
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.V.D.S.); (F.F.); (P.C.); (M.M.); (R.F.)
| |
Collapse
|
14
|
Myasoedov NF, Lyapina LA, Andreeva LA, Grigorieva ME, Obergan TY, Shubina TA. The modern view on the role of glyprolines by metabolic syndrome. Med Res Rev 2020; 41:2823-2840. [DOI: 10.1002/med.21748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 09/17/2020] [Accepted: 10/20/2020] [Indexed: 12/22/2022]
Affiliation(s)
| | | | | | | | - Tamara Y. Obergan
- Department of Biology M. V. Lomonosov Moscow State University Moscow Russia
| | - Tatiana A. Shubina
- Department of Biology M. V. Lomonosov Moscow State University Moscow Russia
| |
Collapse
|
15
|
Ticagrelor Increases SIRT1 and HES1 mRNA Levels in Peripheral Blood Cells from Patients with Stable Coronary Artery Disease and Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2020; 21:ijms21051576. [PMID: 32106619 PMCID: PMC7084534 DOI: 10.3390/ijms21051576] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/21/2020] [Accepted: 02/22/2020] [Indexed: 12/13/2022] Open
Abstract
Ticagrelor is a powerful P2Y12 inhibitor with pleiotropic effects in the cardiovascular system. Consistently, we have reported that in patients with stable coronary artery disease (CAD) and concomitant chronic obstructive pulmonary disease (COPD) who underwent percutaneous coronary intervention (PCI), 1-month treatment with ticagrelor was superior in improving biological markers of endothelial function, compared with clopidogrel. The objective of this study was to investigate the mechanisms underlying these beneficial effects of ticagrelor by conducting molecular analyses of RNA isolated from peripheral blood cells of these patients. We determined mRNAs levels of markers of inflammation and oxidative stress, such as RORγt (T helper 17 cells marker), FoxP3 (regulatory T cells marker), NLRP3, ICAM1, SIRT1, Notch ligands JAG1 and DLL4, and HES1, a Notch target gene. We found that 1-month treatment with ticagrelor, but not clopidogrel, led to increased levels of SIRT1 and HES1 mRNAs. In patients treated with ticagrelor or clopidogrel, we observed a negative correlation among changes in both SIRT1 and HES1 mRNA and serum levels of Epidermal Growth Factor (EGF), a marker of endothelial dysfunction found to be reduced by ticagrelor treatment in our previous study. In conclusion, we report that in stable CAD/COPD patients ticagrelor positively regulates HES1 and SIRT1, two genes playing a protective role in the context of inflammation and oxidative stress. Our observations confirm and expand previous studies showing that the beneficial effects of ticagrelor in stable CAD/COPD patients may be, at least in part, mediated by its capacity to reduce systemic inflammation and oxidative stress.
Collapse
|
16
|
KRIT1 Deficiency Promotes Aortic Endothelial Dysfunction. Int J Mol Sci 2019; 20:ijms20194930. [PMID: 31590384 PMCID: PMC6801783 DOI: 10.3390/ijms20194930] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/20/2019] [Accepted: 09/30/2019] [Indexed: 01/07/2023] Open
Abstract
Loss-of-function mutations of the gene encoding Krev interaction trapped protein 1 (KRIT1) are associated with the pathogenesis of Cerebral Cavernous Malformation (CCM), a major cerebrovascular disease characterized by abnormally enlarged and leaky capillaries and affecting 0.5% of the human population. However, growing evidence demonstrates that KRIT1 is implicated in the modulation of major redox-sensitive signaling pathways and mechanisms involved in adaptive responses to oxidative stress and inflammation, suggesting that its loss-of-function mutations may have pathological effects not limited to CCM disease. The aim of this study was to address whether KRIT1 loss-of-function predisposes to the development of pathological conditions associated with enhanced endothelial cell susceptibility to oxidative stress and inflammation, such as arterial endothelial dysfunction (ED) and atherosclerosis. Silencing of KRIT1 in human aortic endothelial cells (HAECs), coronary artery endothelial cells (HCAECs), and umbilical vein endothelial cells (HUVECs) resulted in increased expression of endothelial proinflammatory adhesion molecules vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) and in enhanced susceptibility to tumor necrosis factor alpha (TNF-α)-induced apoptosis. These effects were associated with a downregulation of Notch1 activation that could be rescued by antioxidant treatment, suggesting that they are consequent to altered intracellular redox homeostasis induced by KRIT1 loss-of-function. Furthermore, analysis of the aorta of heterozygous KRIT1+/- mice fed a high-fructose diet to induce systemic oxidative stress and inflammation demonstrated a 1.6-fold increased expression of VCAM-1 and an approximately 2-fold enhanced fat accumulation (7.5% vs 3.6%) in atherosclerosis-prone regions, including the aortic arch and aortic root, as compared to corresponding wild-type littermates. In conclusion, we found that KRIT1 deficiency promotes ED, suggesting that, besides CCM, KRIT1 may be implicated in genetic susceptibility to the development of atherosclerotic lesions.
Collapse
|
17
|
Aquila G, Kostina A, Vieceli Dalla Sega F, Shlyakhto E, Kostareva A, Marracino L, Ferrari R, Rizzo P, Malaschicheva A. The Notch pathway: a novel therapeutic target for cardiovascular diseases? Expert Opin Ther Targets 2019; 23:695-710. [PMID: 31304807 DOI: 10.1080/14728222.2019.1641198] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: The Notch pathway is involved in determining cell fate during development and postnatally in continuously renewing tissues, such as the endothelium, the epithelium, and in the stem cells pool. The dysregulation of the Notch pathway is one of the causes of limited response, or resistance, to available cancer treatments and novel therapeutic strategies based on Notch inhibition are being investigated in preclinical and clinical studies in oncology. A large body of evidence now shows that the dysregulation of the Notch pathway is also involved in the pathophysiology of cardiovascular diseases (CVDs). Areas covered: This review discusses the molecular mechanisms involving Notch which underlie heart failure, aortic valve calcification, and aortic aneurysm. Expert opinion: Despite the existence of preventive, pharmacological and surgical interventions approaches, CVDs are the first causes of mortality worldwide. The Notch pathway is becoming increasingly recognized as being involved in heart failure, aortic aneurysm and aortic valve calcification, which are among the most common global causes of mortality due to CVDs. As already shown in cancer, the dissection of the biological processes and molecular mechanisms involving Notch should pave the way for new strategies to prevent and cure these diseases.
Collapse
Affiliation(s)
- Giorgio Aquila
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Aleksandra Kostina
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia.,Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences , St-Petersburg , Russia
| | | | - Eugeniy Shlyakhto
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia
| | - Anna Kostareva
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia
| | - Luisa Marracino
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara , Ferrara , Italy
| | - Roberto Ferrari
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy.,Maria Cecilia Hospital, GVM Care & Research , Cotignola , Italy
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research , Cotignola , Italy.,Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara , Ferrara , Italy
| | - Anna Malaschicheva
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia.,Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences , St-Petersburg , Russia.,Department of Embryology, Faculty of Biology, Saint-Petersburg State University , St. Petersburg , Russia
| |
Collapse
|
18
|
Vieceli Dalla Sega F, Fortini F, Aquila G, Campo G, Vaccarezza M, Rizzo P. Notch Signaling Regulates Immune Responses in Atherosclerosis. Front Immunol 2019; 10:1130. [PMID: 31191522 PMCID: PMC6540611 DOI: 10.3389/fimmu.2019.01130] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 05/03/2019] [Indexed: 01/05/2023] Open
Abstract
Atherosclerosis is a chronic autoimmune inflammatory disease that can cause coronary artery disease, stroke, peripheral artery disease, depending on which arteries are affected. At the beginning of atherosclerosis plasma lipoproteins accumulate in the sub-endothelial space. In response, monocytes migrate from the circulation through the endothelium into the intima where they differentiate into macrophages. These early events trigger a complex immune response that eventually involves many cellular subtypes of both innate and adaptive immunity. The Notch signaling pathway is an evolutionary conserved cell signaling system that mediates cell-to-cell communication. Recent studies have revealed that Notch modulate atherosclerosis by controlling macrophages polarization into M1 or M2 subtypes. Furthermore, it is known that Notch signaling controls differentiation and activity of T-helper and cytotoxic T-cells in inflammatory diseases. In this review, we will discuss the role of Notch in modulating immunity in the context of atherosclerosis and whether targeting Notch may represent a therapeutic strategy.
Collapse
Affiliation(s)
| | - Francesca Fortini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, Cotignola, Italy
| | - Giorgio Aquila
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Gianluca Campo
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, Cotignola, Italy.,Cardiovascular Center, Azienda Ospedaliero-Universitaria di Ferrara, Cona, Italy
| | - Mauro Vaccarezza
- Faculty of Health Sciences, School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA, Australia
| | - Paola Rizzo
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, Cotignola, Italy.,Department of Morphology, Surgery, and Experimental Medicine, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| |
Collapse
|
19
|
The Use of Nutraceuticals to Counteract Atherosclerosis: The Role of the Notch Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5470470. [PMID: 31915510 PMCID: PMC6935452 DOI: 10.1155/2019/5470470] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 03/13/2019] [Indexed: 12/13/2022]
Abstract
Despite the currently available pharmacotherapies, today, thirty percent of worldwide deaths are due to cardiovascular diseases (CVDs), whose primary cause is atherosclerosis, an inflammatory disorder characterized by the buildup of lipid deposits on the inside of arteries. Multiple cellular signaling pathways have been shown to be involved in the processes underlying atherosclerosis, and evidence has been accumulating for the crucial role of Notch receptors in regulating the functions of the diverse cell types involved in atherosclerosis onset and progression. Several classes of nutraceuticals have potential benefits for the prevention and treatment of atherosclerosis and CVDs, some of which could in part be due to their ability to modulate the Notch pathway. In this review, we summarize the current state of knowledge on the role of Notch in vascular health and its modulation by nutraceuticals for the prevention of atherosclerosis and/or treatment of related CVDs.
Collapse
|
20
|
Fortini F, Vieceli Dalla Sega F, Caliceti C, Lambertini E, Pannuti A, Peiffer DS, Balla C, Rizzo P. Estrogen-mediated protection against coronary heart disease: The role of the Notch pathway. J Steroid Biochem Mol Biol 2019; 189:87-100. [PMID: 30817989 DOI: 10.1016/j.jsbmb.2019.02.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/05/2019] [Accepted: 02/20/2019] [Indexed: 12/28/2022]
Abstract
Estrogen regulates a plethora of biological processes, under physiological and pathological conditions, by affecting key pathways involved in the regulation of cell proliferation, fate, survival and metabolism. The Notch receptors are mediators of communication between adjacent cells and are key determinants of cell fate during development and in postnatal life. Crosstalk between estrogen and the Notch pathway intervenes in many processes underlying the development and maintenance of the cardiovascular system. The identification of molecular mechanisms underlying the interaction between these types of endocrine and juxtacrine signaling are leading to a deeper understanding of physiological conditions regulated by these steroid hormones and, potentially, to novel therapeutic approaches to prevent pathologies linked to reduced levels of estrogen, such as coronary heart disease, and cardiotoxicity caused by hormone therapy for estrogen-receptor-positive breast cancer.
Collapse
Affiliation(s)
| | | | - Cristiana Caliceti
- Department of Chemistry "Giacomo Ciamician", Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Elisabetta Lambertini
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Antonio Pannuti
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Daniel S Peiffer
- Oncology Research Institute, Loyola University Chicago: Health Sciences Division, Maywood, Illinois, USA; Department of Microbiology and Immunology, Loyola University Chicago: Health Sciences Division, Maywood, Illinois, USA
| | - Cristina Balla
- Cardiovascular Center, University of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, RA, Italy; Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy; Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
21
|
Azuara-Medina PM, Sandoval-Duarte AM, Morales-Lázaro SL, Modragón-González R, Vélez-Aguilera G, Gómez-López JDD, Jiménez-Gutiérrez GE, Tiburcio-Félix R, Martínez-Vieyra I, Suárez-Sánchez R, Längst G, Magaña JJ, Winder SJ, Ortega A, Ramos Perlingeiro RDC, Jacobs LA, Cisneros B. The intracellular domain of β-dystroglycan mediates the nucleolar stress response by suppressing UBF transcriptional activity. Cell Death Dis 2019; 10:196. [PMID: 30814495 PMCID: PMC6393529 DOI: 10.1038/s41419-019-1454-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 01/21/2019] [Accepted: 02/11/2019] [Indexed: 12/12/2022]
Abstract
β-dystroglycan (β-DG) is a key component of multiprotein complexes in the plasma membrane and nuclear envelope. In addition, β-DG undergoes two successive proteolytic cleavages that result in the liberation of its intracellular domain (ICD) into the cytosol and nucleus. However, stimuli-inducing ICD cleavage and the physiological relevance of this proteolytic fragment are largely unknown. In this study we show for the first time that β-DG ICD is targeted to the nucleolus where it interacts with the nuclear proteins B23 and UBF (central factor of Pol I-mediated rRNA gene transcription) and binds to rDNA promoter regions. Interestingly DG silencing results in reduced B23 and UBF levels and aberrant nucleolar morphology. Furthermore, β-DG ICD cleavage is induced by different nucleolar stressors, including oxidative stress, acidosis, and UV irradiation, which implies its participation in the response to nucleolar stress. Consistent with this idea, overexpression of β-DG elicited mislocalization and decreased levels of UBF and suppression of rRNA expression, which in turn provoked altered ribosome profiling and decreased cell growth. Collectively our data reveal that β-DG ICD acts as negative regulator of rDNA transcription by impeding the transcriptional activity of UBF, as a part of the protective mechanism activated in response to nucleolar stress.
Collapse
Affiliation(s)
- Paulina Margarita Azuara-Medina
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional, 07360, Ciudad de México, Mexico
| | - Ariana María Sandoval-Duarte
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional, 07360, Ciudad de México, Mexico
| | - Sara L Morales-Lázaro
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, Mexico
| | - Ricardo Modragón-González
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional, 07360, Ciudad de México, Mexico
| | - Griselda Vélez-Aguilera
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional, 07360, Ciudad de México, Mexico
| | - Juan de Dios Gómez-López
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional, 07360, Ciudad de México, Mexico
| | - Guadalupe Elizabeth Jiménez-Gutiérrez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional, 07360, Ciudad de México, Mexico
| | - Reynaldo Tiburcio-Félix
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional, 07360, Ciudad de México, Mexico
| | - Ivette Martínez-Vieyra
- Laboratorio de Hematobiología, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, 07320, Ciudad de México, Mexico
| | - Rocío Suárez-Sánchez
- Laboratorio de Medicina Genómica, Instituto Nacional de Rehabilitación, 14389, Ciudad de México, Mexico
| | - Gernot Längst
- Biochemistry Centre Regensburg (BCR), Universität Regensburg, 93053, Regensburg, Germany
| | - Jonathan Javier Magaña
- Laboratorio de Medicina Genómica, Instituto Nacional de Rehabilitación, 14389, Ciudad de México, Mexico
| | - Steve J Winder
- Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional, 07000, Ciudad de México, Mexico
| | | | - Laura A Jacobs
- Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK
| | - Bulmaro Cisneros
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional, 07360, Ciudad de México, Mexico.
| |
Collapse
|
22
|
NOTCH1 signaling induces pathological vascular permeability in diabetic retinopathy. Proc Natl Acad Sci U S A 2019; 116:4538-4547. [PMID: 30787185 DOI: 10.1073/pnas.1814711116] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Diabetic macular edema is a major complication of diabetes resulting in loss of central vision. Although heightened vessel leakiness has been linked to glial and neuronal-derived factors, relatively little is known on the mechanisms by which mature endothelial cells exit from a quiescent state and compromise barrier function. Here we report that endothelial NOTCH1 signaling in mature diabetic retinas contributes to increased vascular permeability. By providing both human and mouse data, we show that NOTCH1 ligands JAGGED1 and DELTA LIKE-4 are up-regulated secondary to hyperglycemia and activate both canonical and rapid noncanonical NOTCH1 pathways that ultimately disrupt endothelial adherens junctions in diabetic retinas by causing dissociation of vascular endothelial-cadherin from β-catenin. We further demonstrate that neutralization of NOTCH1 ligands prevents diabetes-induced retinal edema. Collectively, these results identify a fundamental process in diabetes-mediated vascular permeability and provide translational rational for targeting the NOTCH pathway (primarily JAGGED1) in conditions characterized by compromised vascular barrier function.
Collapse
|
23
|
Zhang HM, Chen W, Liu RN, Zhao Y. Notch inhibitor can attenuate apparent diffusion coefficient and improve neurological function through downregulating NOX2-ROS in severe traumatic brain injury. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:3847-3854. [PMID: 30510400 PMCID: PMC6231429 DOI: 10.2147/dddt.s174037] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Introduction Secondary brain injury is a major factor that affects the prognosis and outcome of traumatic brain injury (TBI) patients. Secondary brain edema is considered to be an initiating factor in secondary brain injury after TBI. A previous study has indicated that Notch signaling activation contributes to neuron death in mice affected by stroke; however, its role in neuronal oxidation stress for brain edema after TBI is not well established. Apparent diffusion coefficient (ADC) values can represent the brain edema after TBI. Methods We established a rat model of acute craniocerebral injury, using functional MRI to evaluate the ADC and cerebral blood flow values. The present study was designed to determine the effect of Notch inhibitor DAPT upon oxidation stress for brain edema after TBI. Rats were randomly distributed into five groups, control group, severe TBI group, severe TBI + vehicle group, severe TBI + DAPT group, and severe TBI + DPI group. All rats were sacrificed at 24 hours after TBI. Results Our data indicated that Notch signaling inhibitor DAPT significantly reduced the ADC values and improved the neurological function after TBI. In addition, DAPT decreased NOX2 levels and the ROS levels. Furthermore, DPI can decrease NOX2 levels and ROS levels. Conclusion This study indicated that DAPT Notch signal inhibitors can inhibit NOX2-ROS generation, reduce the ADC values, relieve cerebral edema, and improve nerve function.
Collapse
Affiliation(s)
- Hong-Mei Zhang
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, People's Republic of China, .,Emergency Department, The Second Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, People's Republic of China
| | - Wei Chen
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, People's Republic of China
| | - Rui-Ning Liu
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, People's Republic of China,
| | - Yan Zhao
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, People's Republic of China,
| |
Collapse
|
24
|
Rossini M, Rizzo P, Bononi I, Clementz A, Ferrari R, Martini F, Tognon MG. New Perspectives on Diagnosis and Therapy of Malignant Pleural Mesothelioma. Front Oncol 2018; 8:91. [PMID: 29666782 PMCID: PMC5891579 DOI: 10.3389/fonc.2018.00091] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/15/2018] [Indexed: 12/24/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare, but severe form of cancer, with an incidence that varies significantly within and among different countries around the world. It develops in about one to two persons per million of the general population, leading to thousands of deaths every year worldwide. To date, the MPM is mostly associated with occupational asbestos exposure. Asbestos represents the predominant etiological factor, with approximately 70% of cases of MPM with well-documented occupational exposure to asbestos, with the exposure time, on average greater than 40 years. Environmental exposure to asbestos is increasingly becoming recognized as a cause of mesothelioma, together with gene mutations. The possible roles of other cofactors, such as viral infection and radiation exposure, are still debated. MPM is a fatal tumor. This cancer arises during its early phase without clinical signs. Consequently, its diagnosis occurs at advanced stages. Standard clinical therapeutic approaches include surgery, chemo- and radiotherapies. Preclinical and clinical researches are making great strides in the field of this deadly disease, identifying new biomarkers and innovative therapeutic approaches. Among the newly identified markers and potential therapeutic targets, circulating microRNAs and the Notch pathway represent promising avenues that could result in the early detection of the tumor and novel therapeutic approaches.
Collapse
Affiliation(s)
- Marika Rossini
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Ilaria Bononi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Anthony Clementz
- Department of Natural Sciences and Geography, Concordia University Chicago, River Forest, IL, United States
| | - Roberto Ferrari
- Department of Medical Sciences, Section of Internal Medicine and Cardiorespiratory, School of Medicine, University of Ferrara, Ferrara, Italy.,E.S. Health Science Foundation, GVM Care & Research, Maria Cecilia Hospital, Cotignola, Italy
| | - Fernanda Martini
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Mauro G Tognon
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
25
|
Abstract
Atherosclerosis is a progressive disease of large arteries and a leading cause of cardiovascular diseases and stroke. Chronic inflammation, aberrant immune response, and disturbances to key enzymes involved with lipid metabolism are characteristic features of atherosclerosis. Apart from targeting the derangements in lipid metabolism, therapeutic modulation to regulate chronic inflammation and the immune system response may prove to be very promising strategies in the management of atherosclerosis. In recent years, various targets have been studied for the treatment of atherosclerosis. PCSK9, a serine protease, actively targets the LDL-R and causes lysosomal degradation, which leads to excessive accumulation of LDL-C. Regulatory T cells (Tregs) and Triggering Receptor Expressed on Myeloid cells-1 (TREM-1) affects the adaptive and innate immune response, respectively, and thus, therapeutic intervention of either of these targets would directly modulate disease progression. Advanced atherosclerotic lesions are characterized by an accumulation of apoptotic cells. Cluster of differentiation-47 (CD47), an anti-phagocytic known as the "don't eat me" signaling molecule, inhibits efferocytosis, which causes accumulation of cell debris in plaque. ADAMTS and Notch signaling potentially affect the formation of neointima by modulation of extracellular matrix components such as macrophages and vascular smooth muscle cells. This review provides insights on the molecular targets for therapeutic intervention of atherosclerosis, their effect at various stages of atherosclerosis development, and the therapies that have been designed and currently being evaluated in clinical trials.
Collapse
Affiliation(s)
- Ankita Solanki
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| | - Thomas P Johnston
- Division of Pharmaceutical Sciences, University of Missouri-Kansas City, Kansas City, MO, United States
| |
Collapse
|
26
|
Chen W, Yang J, Chen S, Xiang H, Liu H, Lin D, Zhao S, Peng H, Chen P, Chen AF, Lu H. Importance of mitochondrial calcium uniporter in high glucose-induced endothelial cell dysfunction. Diab Vasc Dis Res 2017; 14:494-501. [PMID: 28777009 PMCID: PMC5652647 DOI: 10.1177/1479164117723270] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE Mitochondrial Ca2+ overload is implicated in hyperglycaemia-induced endothelial cell dysfunction, but the key molecular events responsible remain unclear. We examined the involvement of mitochondrial calcium uniporter, which mediates mitochondrial Ca2+ uptake, in endothelial cell dysfunction resulting from high-glucose treatment. METHODS Human umbilical vein endothelial cells were exposed to various glucose concentrations and to high glucose (30 mM) following mitochondrial calcium uniporter inhibition or activation with ruthenium red and spermine, respectively. Subsequently, mitochondrial calcium uniporter and mitochondrial calcium uniporter regulator 1 messenger RNA and protein expression was measured by real-time polymerase chain reaction and western blotting. Ca2+ concentrations were analysed by laser confocal microscopy, and cytoplasmic and mitochondrial oxidative stress was detected using 2',7'-dichlorofluorescein diacetate and MitoSOX Red, respectively. Apoptosis was assessed by annexin V-fluorescein isothiocyanate/propidium iodide staining, and a wound-healing assay was performed using an in vitro model. RESULTS High glucose markedly upregulated mitochondrial calcium uniporter and mitochondrial calcium uniporter regulator 1 messenger RNA expression, as well as protein production, in a dose- and time-dependent manner with a maximum effect demonstrated at 72 h and 30 mM glucose concentration. Moreover, high-glucose treatment significantly raised both mitochondrial and cytoplasmic Ca2+ and reactive oxygen species levels, increased apoptosis and compromised wound healing (all p < 0.05). These effects were enhanced by spermine and completely negated by ruthenium red, which are known to activate and inhibit mitochondrial calcium uniporter, respectively. CONCLUSION Mitochondrial calcium uniporter plays an important role in hyperglycaemia-induced endothelial cell dysfunction and may constitute a therapeutic target to reduce vascular complications in diabetes.
Collapse
Affiliation(s)
- Wei Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Jie Yang
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Shuhua Chen
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, Hunan, P.R. China
| | - Hong Xiang
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Hengdao Liu
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Dan Lin
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao, Shandong, P.R. China
| | - Shaoli Zhao
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Hui Peng
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Pan Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Alex F Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hongwei Lu
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
- Hongwei Lu, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, P.R. China.
| |
Collapse
|