1
|
Khattib A, Shmet M, Levi A, Hayek T, Halabi M, Khatib S. Bioactive lipids improve serum HDL and PON1 activities in coronary artery disease patients: Ex-vivo study. Vascul Pharmacol 2024; 157:107435. [PMID: 39419293 DOI: 10.1016/j.vph.2024.107435] [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: 09/01/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (CVD) remains a leading cause of vascular disease worldwide. Atherosclerosis is characterized by the accumulation of lipids and oxidized lipids on the blood vessel walls. Coronary artery disease (CAD) is the most common display of atherosclerotic CVD. OBJECTIVES We investigated the effects of the bioactive lipids as lyso-diacylglyceryltrimethylhomoserine (lyso-DGTS (20,5,0)) and its derivative oleoyl-N-trimethyl homoserine amide (oleoyl amide-MHS) on the properties and functionality of HDL and paraoxonase 1 (PON1) activities in the serum of individuals who exhibited arterial plaque as observed by coronary CT angiography (CCTA). METHODS The study included two independent groups comprising 40 patients who had undergone arterial CCTA scans at Ziv Medical Center for various medical indications. The CAD group included 20 patients with coronary artery plaques with luminal stenosis of more than 50 % in a major coronary vessel. The control group consisted of 20 healthy patients (patients without artery plaques). RESULTS Serum samples from CAD patients exhibited lower serum PON1 and cholesterol efflux activities and higher pro-inflammatory than the control group. HDL isolated from CAD patients contains elevated levels of oxidizing lipids (specifically lyso- phosphatidyl ethanolamines and lyso-phosphocholines(compared to the control. However, incubation of the CAD patients' serum with lyso-DGTS and oleoyl amide-MHS restored the antiatherogenic activities of HDL. The lipids increased serum PON1 activities, enhanced apoB-depleted serum cholesterol-efflux activity, and elevated the serum's anti-inflammatory properties. CONCLUSIONS The results of the present study suggest the potential of the bioactive lipids lyso-DGTS and oleoyl amide-MHS to attenuate atherosclerosis via the improvement of dysfunctional HDL properties and PON1 activities. Further, in-vivo experiments are needed to assess the athero-protective effect of the lipids.
Collapse
Affiliation(s)
- Ali Khattib
- Natural Products and Analytical Chemistry Laboratory, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel; Technion Israel Institute of Technology, The Ruth and Bruce Rappaport Faculty of Medicine, Haifa, Israel
| | - Manar Shmet
- Natural Products and Analytical Chemistry Laboratory, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | - Achinoam Levi
- Natural Products and Analytical Chemistry Laboratory, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | - Tony Hayek
- Technion Israel Institute of Technology, The Ruth and Bruce Rappaport Faculty of Medicine, Haifa, Israel
| | | | - Soliman Khatib
- Natural Products and Analytical Chemistry Laboratory, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel.
| |
Collapse
|
2
|
Tang N, Ji L, Shi X, Xiong Y, Xiong X, Zhao H, Song H, Wang J, Zhang L, You S, Ji G, Liu B, Wu N. Effects of Ganjianglingzhu Decoction on Lean Non-Alcoholic Fatty Liver Disease in Mice Based on Untargeted Metabolomics. Pharmaceuticals (Basel) 2024; 17:502. [PMID: 38675462 PMCID: PMC11053674 DOI: 10.3390/ph17040502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/28/2024] [Accepted: 03/31/2024] [Indexed: 04/28/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is usually associated with obesity. However, it is crucial to recognize that NAFLD can also occur in lean individuals, which is frequently overlooked. Without an approved pharmacological therapy for lean NAFLD, we aimed to investigate whether the Ganjianglingzhu (GJLZ) decoction, a representative traditional Chinese medicine (TCM), protects against lean NAFLD and explore the potential mechanism underlying these protective effects. The mouse model of lean NAFLD was established with a methionine-choline-deficient (MCD) diet in male C57BL/6 mice to be compared with the control group fed the methionine-choline-sufficient (MCS) diet. After four weeks, physiological saline, a low dose of GJLZ decoction (GL), or a high dose of GJLZ decoction (GH) was administered daily by gavage to the MCD group; the MCS group was given physiological saline by gavage. Untargeted metabolomics techniques were used to explore further the potential mechanism of the effects of GJLZ on lean NAFLD. Different doses of GJLZ decoction were able to ameliorate steatosis, inflammation, fibrosis, and oxidative stress in the liver; GL performed a better effect on lean NAFLD. In addition, 78 candidate differential metabolites were screened and identified. Combined with metabolite pathway enrichment analysis, GL was capable of regulating the glucose and lipid metabolite pathway in lean NAFLD and regulating the glycerophospholipid metabolism by altering the levels of sn-3-O-(geranylgeranyl)glycerol 1-phosphate and lysoPC(P-18:0/0:0). GJLZ may protect against the development of lean NAFLD by regulating glucose and lipid metabolism, inhibiting the levels of sn-3-O-(geranylgeranyl)glycerol 1-phosphate and lysoPC(P-18:0/0:0) in glycerophospholipid metabolism.
Collapse
Affiliation(s)
- Nan Tang
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (N.T.); (X.S.); (Y.X.); (X.X.); (H.S.); (J.W.); (L.Z.); (G.J.)
| | - Lei Ji
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai 200030, China;
| | - Xinyu Shi
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (N.T.); (X.S.); (Y.X.); (X.X.); (H.S.); (J.W.); (L.Z.); (G.J.)
| | - Yalan Xiong
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (N.T.); (X.S.); (Y.X.); (X.X.); (H.S.); (J.W.); (L.Z.); (G.J.)
| | - Xinying Xiong
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (N.T.); (X.S.); (Y.X.); (X.X.); (H.S.); (J.W.); (L.Z.); (G.J.)
| | - Hanhua Zhao
- Department of Sport Science, College of Education, Zhejiang University, Hangzhou 310058, China;
| | - Hualing Song
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (N.T.); (X.S.); (Y.X.); (X.X.); (H.S.); (J.W.); (L.Z.); (G.J.)
| | - Jianying Wang
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (N.T.); (X.S.); (Y.X.); (X.X.); (H.S.); (J.W.); (L.Z.); (G.J.)
| | - Lei Zhang
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (N.T.); (X.S.); (Y.X.); (X.X.); (H.S.); (J.W.); (L.Z.); (G.J.)
| | - Shengfu You
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China;
| | - Guang Ji
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (N.T.); (X.S.); (Y.X.); (X.X.); (H.S.); (J.W.); (L.Z.); (G.J.)
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China;
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Baocheng Liu
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (N.T.); (X.S.); (Y.X.); (X.X.); (H.S.); (J.W.); (L.Z.); (G.J.)
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Na Wu
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (N.T.); (X.S.); (Y.X.); (X.X.); (H.S.); (J.W.); (L.Z.); (G.J.)
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
3
|
Deshpande N, Wilkins MR, Khachigian LM. RNA sequencing identifies genes reliant upon Ser26 in early growth response-1 in vascular endothelial cells exposed to fibroblast growth factor-2. Vascul Pharmacol 2022; 143:106952. [PMID: 35041980 DOI: 10.1016/j.vph.2022.106952] [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: 12/17/2021] [Accepted: 01/08/2022] [Indexed: 11/25/2022]
Abstract
Early growth response-1 (Egr-1) is an inducible master regulatory transcription factor that orchestrates gene expression in vascular endothelial cells. We recently determined that Ser26 in Egr-1 undergoes phosphorylation and plays a critical functional role in a range of pro-angiogenic processes. To better understand the effect of Ser26 on Egr-1-dependent gene expression, in this study, we performed RNA-seq and bioinformatics analysis on human microvascular endothelial cells bearing a germline mutation (M) in Ser26 to Ala (M26 cells) exposed to the mitogen and chemoattractant fibroblast growth factor-2 (FGF2) as compared with wildtype (WT) cells. In WT cells, FGF2 increased the expression of numerous growth factors and hormones cytokines, signaling molecules and transcriptional regulators. Comparison of FGF2-inducible WT and M26 cells enabled identification of differentially expressed genes, including genes reliant or not reliant upon Ser26. For example, Ser26 in Egr-1 was required for FGF2 inducible LIF expression but not for FGF2 inducible IL11. Ser26 was also required for FGF2 inducible NKX2-8 and RIPK2 expression but not for FGF2 inducible CREB5 or ALPK2 expression. Conversely, FGF2 inhibited genes such as TIE1, GPR146 and EPHB3, and Ser26 was required for FGF2's effect on TIE1 and GPR146 but not for EPHB3. Enrichment analysis also identified a range of gene ontologies upregulated and downregulated by FGF2. These findings demonstrate the importance of Ser26 in Egr-1 in programs of endothelial gene expression modulated by FGF2.
Collapse
Affiliation(s)
- Nandan Deshpande
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Marc R Wilkins
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research, School of Medical Sciences & UNSW Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
4
|
Cabrera JTO, Makino A. Efferocytosis of vascular cells in cardiovascular disease. Pharmacol Ther 2022; 229:107919. [PMID: 34171333 PMCID: PMC8695637 DOI: 10.1016/j.pharmthera.2021.107919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/21/2021] [Accepted: 06/03/2021] [Indexed: 12/20/2022]
Abstract
Cell death and the clearance of apoptotic cells are tightly regulated by various signaling molecules in order to maintain physiological tissue function and homeostasis. The phagocytic removal of apoptotic cells is known as the process of efferocytosis, and abnormal efferocytosis is linked to various health complications and diseases, such as cardiovascular disease, inflammatory diseases, and autoimmune diseases. During efferocytosis, phagocytic cells and/or apoptotic cells release signals, such as "find me" and "eat me" signals, to stimulate the phagocytic engulfment of apoptotic cells. Primary phagocytic cells are macrophages and dendritic cells; however, more recently, other neighboring cell types have also been shown to exhibit phagocytic character, including endothelial cells and fibroblasts, although they are comparatively slower in clearing dead cells. In this review, we focus on macrophage efferocytosis of vascular cells, such as endothelial cells, smooth muscle cells, fibroblasts, and pericytes, and its relation to the progression and development of cardiovascular disease. We also highlight the role of efferocytosis-related molecules and their contribution to the maintenance of vascular homeostasis.
Collapse
Affiliation(s)
- Jody Tori O Cabrera
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
5
|
Zhao H, He Y. The Inhibitory Effect of Lysophosphatidylcholine on Proangiogenesis of Human CD34 + Cells Derived Endothelial Progenitor Cells. Front Mol Biosci 2021; 8:682367. [PMID: 34179086 PMCID: PMC8223510 DOI: 10.3389/fmolb.2021.682367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/28/2021] [Indexed: 12/27/2022] Open
Abstract
Increasing evidence reveals that lysophosphatidylcholine (LPC) is closely related to endothelial dysfunction. The present study aimed to investigate the mechanism of LPC in inhibiting the proangiogenesis and vascular inflammation of human endothelial progenitor cells (EPCs) derived from CD34+ cells. The early EPCs were derived from CD34+ hematopoietic stem cells whose purity was identified using flow cytometry analysis. The surface markers (CD34, KDR, CD31; VE-cadherin, vWF, eNOS) of EPCs were examined by flow cytometry analysis and immunofluorescence. RT-qPCR was used to detect the mRNA expression of inflammatory cytokines (CCL2, IL-8, CCL4) and genes associated with angiogenesis (VEGF, ANG-1, ANG-2) in early EPCs after treatment of LPC (10 μg/ml) or phosphatidylcholine (PC, 10 μg/ml, control). The angiogenesis of human umbilical vein endothelial cells (HUVECs) incubated with the supernatants of early EPCs was detected by a tube formation assay. The mRNA and protein levels of key factors on the PKC pathway (phosphorylated PKC, TGF-β1) were measured by RT-qPCR and western blot. The localization of PKC-β1 in EPCs was determined by immunofluorescence staining. We found that LPC suppressed the expression of CCL2, CCL4, ANG-1, ANG-2, promoted IL-8 expression and had no significant effects on VEGF expression in EPCs. EPCs promoted the angiogenesis of HUVECs, which was significantly inhibited by LPC treatment. Moreover, LPC was demonstrated to promote the activation of the PKC signaling pathway in EPCs. In conclusion, LPC inhibits proangiogenesis of human endothelial progenitor cells derived from CD34+ hematopoietic stem cells.
Collapse
Affiliation(s)
- Haijun Zhao
- Department of Pain, The First Hospital of Jilin University, Changchun, China
| | - Yanhui He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Li C, Wen R, Liu DW, Liu Q, Yan LP, Wu JX, Guo YJ, Li SY, Gong QF, Yu H. Diuretic Effect and Metabolomics Analysis of Crude and Salt-Processed Plantaginis Semen. Front Pharmacol 2021; 11:563157. [PMID: 33390941 PMCID: PMC7774519 DOI: 10.3389/fphar.2020.563157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/08/2020] [Indexed: 11/13/2022] Open
Abstract
Plantaginis Semen (PS) is well recognized in traditional Chinese medicine (TCM) and health products. Crude PS (CPS) and salt-processed CPS (SPS) are the two most commonly used decoction pieces of PS, and are included in the 2020 edition of Chinese Pharmacopoeia. Although they all have multiple effects, the mechanisms for treating diseases are different and remain unclear, the processing mechanism of SPS is also indeterminate, which hinders their clinical application to a certain extent. In order to solve these problems and further develop PS in the clinical application. Here, we used saline-loaded model rats for experiments, and utilized an integrated approach consisting of pharmacological methods and metabolomics, which could assess the diuretic impact of CPS and SPS ethanol extracts on saline-loaded rats and elucidate the underlying mechanism. The results showed that CPS and SPS both produced increased urine volume excretion and urine electrolyte excretion, but the levels of aldosterone (ALD) and aquaporin 2 (AQP2) were decreased. And 30 differential metabolites such as linoleic acid, lysoPC(O-18:0), sphingosine-1-phosphate, lysoPC(18:0) were found, mainly involving three metabolic pathways. In conclusion, CPS and SPS both have a diuretic effect, and that of SPS is better. This work investigated the possible diuretic mechanisms of CPS and SPS which may also be the mechanism of PS for anti-hypertension. In addition, a holistic approach provided novel and helpful insights into the underlying processing mechanisms of TCM.
Collapse
Affiliation(s)
- Chao Li
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Rou Wen
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - De Wen Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiang Liu
- Department of Chemistry, Stanford University, CA, United States
| | - Li Ping Yan
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Jian Xiong Wu
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yi Jing Guo
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Su Yun Li
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qian Feng Gong
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Huan Yu
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
7
|
Tang X, Wang JJ, Wang J, Abboud HE, Chen Y, Zhang SX. Endothelium-specific deletion of Nox4 delays retinal vascular development and mitigates pathological angiogenesis. Angiogenesis 2020; 24:363-377. [PMID: 33201372 DOI: 10.1007/s10456-020-09757-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 11/06/2020] [Indexed: 11/29/2022]
Abstract
NADPH oxidase 4 (Nox4) is a major isoform of NADPH oxidases playing an important role in many biological processes. Previously we have shown that Nox4 is highly expressed in retinal blood vessels and is upregulated in oxygen-induced retinopathy (OIR). However, the exact role of endothelial Nox4 in retinal angiogenesis remains elusive. Herein, using endothelial cell (EC)-specific Nox4 knockout (Nox4EC-KO) mice, we investigated the impact of endothelial Nox4 deletion on retinal vascular development and pathological angiogenesis during OIR. Our results show that deletion of Nox4 in ECs led to retarded retinal vasculature development with fewer, blunted-end tip cells and sparser, dysmorphic filopodia at vascular front, and reduced density of vascular network in superficial, deep, and intermediate layers in postnatal day 7 (P7), P12, and P17 retinas, respectively. In OIR, loss of endothelial Nox4 had no effect on hyperoxia-induced retinal vaso-obliteration at P9 but significantly reduced aberrant retinal neovascularization at P17 and decreased the deep layer capillary density at P25. Ex vivo study confirmed that lack of Nox4 in ECs impaired vascular sprouting. Mechanistically, loss of Nox4 significantly reduced expression of VEGF, p-VEGFR2, integrin αV, angiopoietin-2, and p-ERK1/2, attenuating EC migration and proliferation. Taken together, our results indicate that endothelial Nox4 is important for retinal vascular development and contributes to pathological angiogenesis, likely through regulation of VEGF/VEGFR2 and angiopoietin-2/integrin αV/ERK pathways. In addition, our study suggests that endothelial Nox4 appears to be essential for intraretinal revascularization after hypoxia. These findings call for caution on targeting endothelial Nox4 in ischemic/hypoxic retinal diseases.
Collapse
Affiliation(s)
- Xixiang Tang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY, USA.,SUNY Eye Institute, State University of New York, New York, NY, USA.,Department of Endocrinology and Metabolism, The Third Affiliated Hospital, Sun Yat-Sen University; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China.,VIP Medical Service Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Joshua J Wang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY, USA.,SUNY Eye Institute, State University of New York, New York, NY, USA
| | - Jinli Wang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY, USA.,SUNY Eye Institute, State University of New York, New York, NY, USA
| | - Hanna E Abboud
- Department of Medicine, South Texas Veterans Healthcare System and the University of Texas Health Science Center, San Antonio, TX, USA
| | - Yanming Chen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital, Sun Yat-Sen University; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Sarah X Zhang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY, USA. .,SUNY Eye Institute, State University of New York, New York, NY, USA. .,Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, NY, USA.
| |
Collapse
|
8
|
Chu CS, Law SH, Lenzen D, Tan YH, Weng SF, Ito E, Wu JC, Chen CH, Chan HC, Ke LY. Clinical Significance of Electronegative Low-Density Lipoprotein Cholesterol in Atherothrombosis. Biomedicines 2020; 8:biomedicines8080254. [PMID: 32751498 PMCID: PMC7460408 DOI: 10.3390/biomedicines8080254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/25/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the numerous risk factors for atherosclerotic cardiovascular diseases (ASCVD), cumulative evidence shows that electronegative low-density lipoprotein (L5 LDL) cholesterol is a promising biomarker. Its toxicity may contribute to atherothrombotic events. Notably, plasma L5 LDL levels positively correlate with the increasing severity of cardiovascular diseases. In contrast, traditional markers such as LDL-cholesterol and triglyceride are the therapeutic goals in secondary prevention for ASCVD, but that is controversial in primary prevention for patients with low risk. In this review, we point out the clinical significance and pathophysiological mechanisms of L5 LDL, and the clinical applications of L5 LDL levels in ASCVD can be confidently addressed. Based on the previously defined cut-off value by receiver operating characteristic curve, the acceptable physiological range of L5 concentration is proposed to be below 1.7 mg/dL. When L5 LDL level surpass this threshold, clinically relevant ASCVD might be present, and further exams such as carotid intima-media thickness, pulse wave velocity, exercise stress test, or multidetector computed tomography are required. Notably, the ultimate goal of L5 LDL concentration is lower than 1.7 mg/dL. Instead, with L5 LDL greater than 1.7 mg/dL, lipid-lowering treatment may be required, including statin, ezetimibe or PCSK9 inhibitor, regardless of the low-density lipoprotein cholesterol (LDL-C) level. Since L5 LDL could be a promising biomarker, we propose that a high throughput, clinically feasible methodology is urgently required not only for conducting a prospective, large population study but for developing therapeutics strategies to decrease L5 LDL in the blood.
Collapse
Affiliation(s)
- Chih-Sheng Chu
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan;
- Division of Cardiology, Department of International Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Shi Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
| | - David Lenzen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
| | - Yong-Hong Tan
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
| | - Shih-Feng Weng
- Department of Healthcare Administration and Medical Informatics, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
| | - Etsuro Ito
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
- Department of Biology, Waseda University, Tokyo 162-8480, Japan
- Waseda Research Institute for Science and Engineering, Waseda University, Tokyo 162-8480, Japan
| | - Jung-Chou Wu
- Division of Cardiology, Department of Internal Medicine, Pingtung Christian Hospital, Pingtung 90059, Taiwan;
| | - Chu-Huang Chen
- Vascular and Medicinal Research, Texas Heart Institute, Houston, TX 77030, USA;
| | - Hua-Chen Chan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan;
- Correspondence: (H.-C.C.); (L.-Y.K.); Tel.: +886-73121101 (ext. 2296); Fax: +886-73111996 (L.-Y.K.)
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
- Graduate Institute of Medicine, College of Medicine, & Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Correspondence: (H.-C.C.); (L.-Y.K.); Tel.: +886-73121101 (ext. 2296); Fax: +886-73111996 (L.-Y.K.)
| |
Collapse
|
9
|
Li BB, Chen ZY, Jiang N, Guo S, Yang JQ, Chai SB, Yan HF, Sun PM, Hu G, Zhang T, Xu BX, Sun HW, Zhou JL, Yang HM, Cui Y. Simulated microgravity significantly altered metabolism in epidermal stem cells. In Vitro Cell Dev Biol Anim 2020; 56:200-212. [PMID: 32198676 PMCID: PMC7186248 DOI: 10.1007/s11626-020-00435-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/08/2020] [Indexed: 12/23/2022]
Abstract
Simulated microgravity can significantly affect various cell types and multiple systems of the human body, such as cardiovascular system, skeletal muscle system, and immune system, and is known to cause anemia and loss of electrolyte and fluids. Epidermal stem cells (EpSCs) were cultured in a rotary cell culture system (RCCS) bioreactor to simulate microgravity. The metabolites of EpSCs were identified by liquid chromatography-mass spectrometry (LC-MS). Compared with normal gravity (NG) group, a total of 57 different metabolites of EpSCs were identified (P < 0.05, VIP > 1), including lipids and lipid-like molecules (51 molecules), amino acids (5 molecules), nucleosides, nucleotides, and analogues (1 molecule). According to the partial least squares discriminant analysis (PLS-DA) score plot, a VIP > 1 and P < 0.05 were obtained for the 57 different metabolites, of which 23 molecules were significantly downregulated and 34 were significantly upregulated in simulated microgravity (SMG) group. These results showed that SMG has a significant impact on different pathways, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis indicated that multiple pathways were involved, mainly the amino acid metabolism pathway, lipid metabolism pathway, membrane transport pathway, and cell growth and death pathways. Thus, the metabolic profile of EpSCs was changed under SMG. Exploring the metabolic profile of EpSCs would be helpful to further understand the growth characteristics of EpSCs under SMG, which will provide a new approach to explore the metabolomics mechanism of stress injury and repair trauma under SMG.
Collapse
Affiliation(s)
- Bin-Bin Li
- Department of General Surgery, PLA 306 Clinical Hospital of Anhui Medical University, Beijing, 230000 China
| | - Zheng-Yang Chen
- Department of General Surgery, PLA 306 Teaching Hospital of Peking University Health Science Center, Beijing, 100101 China
| | - Nan Jiang
- The Center for Hepatopancreatobiliary Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218 China
| | - Song Guo
- Department of General Surgery, PLA 306 Teaching Hospital of Peking University Health Science Center, Beijing, 100101 China
| | - Jia-Qi Yang
- Department of General Surgery, PLA 306 Teaching Hospital of Peking University Health Science Center, Beijing, 100101 China
| | - Shao-Bin Chai
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Hong-Feng Yan
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Pei-Ming Sun
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Gang Hu
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Tao Zhang
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Bing-Xin Xu
- Medicine and Experimental Research Center, 306 Hospital of PLA, Beijing, 100101 China
| | - Hong-Wei Sun
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Jin-Lian Zhou
- Department of Pathology, 306 Hospital of PLA, Beijing, 100101 China
| | - He-Ming Yang
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Yan Cui
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| |
Collapse
|
10
|
Law SH, Chan ML, Marathe GK, Parveen F, Chen CH, Ke LY. An Updated Review of Lysophosphatidylcholine Metabolism in Human Diseases. Int J Mol Sci 2019; 20:ijms20051149. [PMID: 30845751 PMCID: PMC6429061 DOI: 10.3390/ijms20051149] [Citation(s) in RCA: 463] [Impact Index Per Article: 77.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/12/2022] Open
Abstract
Lysophosphatidylcholine (LPC) is increasingly recognized as a key marker/factor positively associated with cardiovascular and neurodegenerative diseases. However, findings from recent clinical lipidomic studies of LPC have been controversial. A key issue is the complexity of the enzymatic cascade involved in LPC metabolism. Here, we address the coordination of these enzymes and the derangement that may disrupt LPC homeostasis, leading to metabolic disorders. LPC is mainly derived from the turnover of phosphatidylcholine (PC) in the circulation by phospholipase A2 (PLA2). In the presence of Acyl-CoA, lysophosphatidylcholine acyltransferase (LPCAT) converts LPC to PC, which rapidly gets recycled by the Lands cycle. However, overexpression or enhanced activity of PLA2 increases the LPC content in modified low-density lipoprotein (LDL) and oxidized LDL, which play significant roles in the development of atherosclerotic plaques and endothelial dysfunction. The intracellular enzyme LPCAT cannot directly remove LPC from circulation. Hydrolysis of LPC by autotaxin, an enzyme with lysophospholipase D activity, generates lysophosphatidic acid, which is highly associated with cancers. Although enzymes with lysophospholipase A1 activity could theoretically degrade LPC into harmless metabolites, they have not been found in the circulation. In conclusion, understanding enzyme kinetics and LPC metabolism may help identify novel therapeutic targets in LPC-associated diseases.
Collapse
Affiliation(s)
- Shi-Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Mei-Lin Chan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
- Division of Thoracic Surgery, Department of Surgery, MacKay Memorial Hospital, MacKay Medical College, Taipei 10449, Taiwan.
| | - Gopal K Marathe
- Department of Studies in Biochemistry, Manasagangothri, University of Mysore, Mysore-570006, India.
| | - Farzana Parveen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Chu-Huang Chen
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Vascular and Medicinal Research, Texas Heart Institute, Houston, TX 77030, USA.
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
11
|
Song M, Finley SD. Mechanistic insight into activation of MAPK signaling by pro-angiogenic factors. BMC SYSTEMS BIOLOGY 2018; 12:145. [PMID: 30591051 PMCID: PMC6307205 DOI: 10.1186/s12918-018-0668-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/30/2018] [Indexed: 01/14/2023]
Abstract
Background Angiogenesis is important in physiological and pathological conditions, as blood vessels provide nutrients and oxygen needed for tissue growth and survival. Therefore, targeting angiogenesis is a prominent strategy in both tissue engineering and cancer treatment. However, not all of the approaches to promote or inhibit angiogenesis lead to successful outcomes. Angiogenesis-based therapies primarily target pro-angiogenic factors such as vascular endothelial growth factor-A (VEGF) or fibroblast growth factor (FGF) in isolation. However, pre-clinical and clinical evidence shows these therapies often have limited effects. To improve therapeutic strategies, including targeting FGF and VEGF in combination, we need a quantitative understanding of the how the promoters combine to stimulate angiogenesis. Results In this study, we trained and validated a detailed mathematical model to quantitatively characterize the crosstalk of FGF and VEGF intracellular signaling. This signaling is initiated by FGF binding to the FGF receptor 1 (FGFR1) and heparan sulfate glycosaminoglycans (HSGAGs) or VEGF binding to VEGF receptor 2 (VEGFR2) to promote downstream signaling. The model focuses on FGF- and VEGF-induced mitogen-activated protein kinase (MAPK) signaling and phosphorylation of extracellular regulated kinase (ERK), which promotes cell proliferation. We apply the model to predict the dynamics of phosphorylated ERK (pERK) in response to the stimulation by FGF and VEGF individually and in combination. The model predicts that FGF and VEGF have differential effects on pERK. Additionally, since VEGFR2 upregulation has been observed in pathological conditions, we apply the model to investigate the effects of VEGFR2 density and trafficking parameters. The model predictions show that these parameters significantly influence the response to VEGF stimulation. Conclusions The model agrees with experimental data and is a framework to synthesize and quantitatively explain experimental studies. Ultimately, the model provides mechanistic insight into FGF and VEGF interactions needed to identify potential targets for pro- or anti-angiogenic therapies. Electronic supplementary material The online version of this article (10.1186/s12918-018-0668-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Min Song
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Stacey D Finley
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA. .,Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA. .,Department of Biological Sciences, Computational Biology section, University of Southern California, 1042 Downey Way, CRB 140, Los Angeles, CA, 90089, USA.
| |
Collapse
|
12
|
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a member of the nuclear receptor family and plays an important role in adipocyte differentiation, glucose homeostasis, and insulin sensitivity. Thiazolidinediones (TZDs), synthetic ligands of PPARγ, have been used for the treatment of diabetes mellitus for two decades. TZDs were expected to be amazing drugs not only for type 2 diabetes but also for metabolic syndrome and atherosclerotic vascular disease because they can reduce both insulin resistance and inflammation in experimental studies. However, serious unwanted effects pushed TZDs back to an optional second-tier drug for type 2 diabetes. Nevertheless, PPARγ is still one of the most important targets for the treatment of insulin resistance and diabetes mellitus, and novel strategies to modulate PPARγ activity to enhance its beneficial effects and reduce unwanted adverse effects are anticipated. Recent studies showed that post-translational modification (PTM) of PPARγ regulates PPARγ activity or stability and may be a novel way to optimize PPARγ activity with reduced adverse effects. In this review, we will focus on recent advances in PTM of PPARγ and the mechanisms regulating PPARγ function as well as in the development of PPARγ modulators or agonists.
Collapse
Affiliation(s)
- Sung Hee Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea.,Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Sung Soo Chung
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| |
Collapse
|
13
|
Tian Y, Jiang F, Li Y, Jiang H, Chu Y, Zhu L, Guo W. Evaluation of the anti-hypertensive effect of Tengfu Jiangya tablet by combination of UPLC-Q-exactive-MS-based metabolomics and iTRAQ-based proteomics technology. Biomed Pharmacother 2018; 100:324-334. [DOI: 10.1016/j.biopha.2018.02.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/07/2018] [Accepted: 02/09/2018] [Indexed: 12/11/2022] Open
|
14
|
Smith NJ, Fuller M, Saville JT, Cox TM. Reduced cerebral vascularization in experimental neuronopathic Gaucher disease. J Pathol 2018; 244:120-128. [PMID: 28981147 DOI: 10.1002/path.4992] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/15/2017] [Accepted: 09/12/2017] [Indexed: 11/10/2022]
Abstract
The glycosphingolipidosis, Gaucher disease, in which a range of neurological manifestations occur, results from a deficiency of acid β-glucocerebrosidase, with subsequent accumulation of β-glucocerebroside, its upstream substrates, and the non-acylated congener β-glucosylsphingosine. However, the mechanisms by which end-organ dysfunction arise are poorly understood. Here, we report strikingly diminished cerebral microvascular density in a murine model of disease, and provide a detailed analysis of the accompanying cerebral glycosphingolipidome in these animals, with marked elevations of β-glucosylsphingosine. Further in vitro studies confirmed a concentration-dependent impairment of endothelial cytokinesis upon exposure to quasi-pathological concentrations of β-glucosylsphingosine. These findings support a premise for pathogenic disruption of cerebral angiogenesis as an end-organ effect, with potential for therapeutic modulation in neuronopathic Gaucher disease. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Nicholas Jc Smith
- Department of Neurology and Clinical Neurophysiology, Women's and Children's Health Network, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia.,Department of Medicine, University of Cambridge, Cambridge, UK
| | - Maria Fuller
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia.,Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, Adelaide, South Australia, Australia
| | - Jennifer T Saville
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, Adelaide, South Australia, Australia
| | - Timothy M Cox
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
15
|
Ku YH, Cho BJ, Kim MJ, Lim S, Park YJ, Jang HC, Choi SH. Rosiglitazone increases endothelial cell migration and vascular permeability through Akt phosphorylation. BMC Pharmacol Toxicol 2017; 18:62. [PMID: 28854981 PMCID: PMC5577739 DOI: 10.1186/s40360-017-0169-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 08/11/2017] [Indexed: 02/06/2023] Open
Abstract
Background Thiazolidinediones (TZDs), peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists, exhibit anti-inflammatory and antioxidant properties and inhibit endothelial inflammation and dysfunction, which is anti-atherogenic. However, fluid retention, which may lead to congestive heart failure and peripheral edema, is also a concern, which may result from endothelial cell leakage. In the current study, we examined the effects of PPAR-γ agonists on vascular endothelial cell migration and permeability in order to determine its underlying mechanisms. Methods We used rosiglitazone and conducted cell migration assay and permeability assay using HUVEC cells and measured vascular permeability and leakage in male C57BL/6 mice. Results Rosiglitazone significantly promoted endothelial cell migration and induced permeability via activation of phosphatidylinositol-3-kinase (PI3K) – Akt or protein kinase C (PKC)β. In addition, rosiglitazone increased vascular endothelial growth factor (VEGF) expression and suppressed expression of tight junction proteins (JAM-A and ZO-1), which might promote neovascularization and vascular leakage. These phenomena were reduced by Akt inhibition. Conclusions Vascular endothelial cell migration and permeability change through Akt phosphorylation might be a mechanism of induced fluid retention and peripheral tissue edema by TZD.
Collapse
Affiliation(s)
- Yun Hyi Ku
- Department of Internal Medicine, Korea Cancer Center Hospital, Seoul, South Korea
| | - Bong-Jun Cho
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Min Joo Kim
- Department of Internal Medicine, Korea Cancer Center Hospital, Seoul, South Korea
| | - Soo Lim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Hak C Jang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Sung Hee Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea. .,, 166 Gumi-dong, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-707, Republic of Korea.
| |
Collapse
|
16
|
Qian LQ, Li XQ, Ye PH, Su HY, Wang G, Liu Y, Shen GH, Gao QG. Downregulation of MACC1 inhibits the viability, invasion and migration and induces apoptosis in esophageal carcinoma cells through the phosphatase and tensin homolog/phosphoinositide 3-kinase/protein kinase B signaling pathway. Oncol Lett 2017; 14:4897-4905. [PMID: 29085498 DOI: 10.3892/ol.2017.6790] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 06/02/2017] [Indexed: 12/12/2022] Open
Abstract
As an oncogene, MACC1 serves an important function in cancer progression and metastasis. However, the effect of MACC1 in esophageal carcinoma (EC) remains to be fully understood. The present study assessed the association between MACC1 expression and the progression of EC cells. A small interfering (si)RNA was delivered into EC cells to downregulate MACC1 expression. The MTT assay demonstrated that EC cell viability was reduced by siRNA-MACC1. Decreasing MACC1 expression increased the apoptotic rate of EC cells compared with control cells. Transwell and Matrigel assays demonstrated that EC cell migration and invasion, respectively, were downregulated by siRNA-MACC1. Furthermore, knocking down MACC1 suppressed the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway by upregulating the expression of phosphatase and tensin homolog (PTEN), a tumor suppressor. The results of the present study revealed that MACC1 expression affected cellular functions of the EC cells through the PTEN/PI3K/Akt signaling pathway. Therefore, MACC1 may potentially serve as a novel biomarker and therapeutic target for EC.
Collapse
Affiliation(s)
- Li-Qiang Qian
- Department of General Surgery, Wujiang No. 1 People's Hospital, Suzhou, Jiangsu 215200, P.R. China
| | - Xia-Qin Li
- Department of Gynaecology and Obstetrics, Health Center of Songling, Suzhou, Jiangsu 215200, P.R. China
| | - Peng-Hui Ye
- Department of General Surgery, Wujiang No. 1 People's Hospital, Suzhou, Jiangsu 215200, P.R. China
| | - Hao-Yuan Su
- Department of General Surgery, Wujiang No. 1 People's Hospital, Suzhou, Jiangsu 215200, P.R. China
| | - Gang Wang
- Department of General Surgery, Wujiang No. 1 People's Hospital, Suzhou, Jiangsu 215200, P.R. China
| | - Yan Liu
- Department of General Surgery, Wujiang No. 1 People's Hospital, Suzhou, Jiangsu 215200, P.R. China
| | - Gen-Hai Shen
- Department of General Surgery, Wujiang No. 1 People's Hospital, Suzhou, Jiangsu 215200, P.R. China
| | - Quan-Gen Gao
- Department of General Surgery, Wujiang No. 1 People's Hospital, Suzhou, Jiangsu 215200, P.R. China
| |
Collapse
|
17
|
C1q/TNF-Related Protein-9 Ameliorates Ox-LDL-Induced Endothelial Dysfunction via PGC-1α/AMPK-Mediated Antioxidant Enzyme Induction. Int J Mol Sci 2017; 18:ijms18061097. [PMID: 28587104 PMCID: PMC5485929 DOI: 10.3390/ijms18061097] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 05/16/2017] [Accepted: 05/17/2017] [Indexed: 02/03/2023] Open
Abstract
Oxidized low-density lipoprotein (ox-LDL) accumulation is one of the critical determinants in endothelial dysfunction in many cardiovascular diseases such as atherosclerosis. C1q/TNF-related protein 9 (CTRP9) is identified to be an adipocytokine with cardioprotective properties. However, the potential roles of CTRP9 in endothelial function remain largely elusive. In the present study, the effects of CTRP9 on the proliferation, apoptosis, migration, angiogenesis, nitric oxide (NO) production and oxidative stress in human umbilical vein endothelial cells (HUVECs) exposed to ox-LDL were investigated. We observed that treatment with ox-LDL inhibited the proliferation, migration, angiogenesis and the generation of NO, while stimulated the apoptosis and reactive oxygen species (ROS) production in HUVECs. Incubation of HUVECs with CTRP9 rescued ox-LDL-induced endothelial injury. CTRP9 treatment reversed ox-LDL-evoked decreases in antioxidant enzymes including heme oxygenase-1 (HO-1), nicotinamide adenine dinucleotide phosphate (NAD(P)H) dehydrogenase quinone 1, and glutamate-cysteine ligase (GCL), as well as endothelial nitric oxide synthase (eNOS). Furthermore, CTRP9 induced activation of peroxisome proliferator-activated receptor γ co-activator 1α (PGC1-α) and phosphorylation of adenosine monophosphate-activated protein kinase (AMPK). Of interest, AMPK inhibition or PGC1-α silencing abolished CTRP9-mediated antioxidant enzymes levels, eNOS expressions, and endothelial protective effects. Collectively, we provided the first evidence that CTRP9 attenuated ox-LDL-induced endothelial injury by antioxidant enzyme inductions dependent on PGC-1α/AMPK activation.
Collapse
|
18
|
QGQS Granule in SHR Serum Metabonomics Study Based on Tools of UPLC-Q-TOF and Renin-Angiotensin-Aldosterone System Form Protein Profilin-1. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:4854720. [PMID: 28367224 PMCID: PMC5358465 DOI: 10.1155/2017/4854720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 01/15/2017] [Accepted: 01/24/2017] [Indexed: 01/09/2023]
Abstract
QGQS granule is effective for the therapeutic of hypertension in clinic. The aim of this research is to observe the antihypertension effect of QGQS granule on SHR and explain the mechanism of its lowering blood pressure. 30 SHR were selected as model group, captopril group, and QGQS group, 10 WKYr were used as control group, and RBP were measured on tail artery consciously. And all the serum sample analysis was carried out on UPLC-TOF-MS system to determine endogenous metabolites and to find the metabonomics pathways. Meanwhile, ELISA kits for the determination pharmacological indexes of PRA, AngI, AngII, and ALD were used for pathway confirmatory; WB for determination of profilin-1 protein expression was conducted for Ang II pathway analysis as well. It is demonstrated that QGQS granule has an excellent therapeutic effect on antihypertension, which exerts effect mainly on metabonomics pathway by regulating glycerophospholipid, sphingolipid, and arachidonic acid metabolism, and it could inhibit the overexpression of the profilin-1 protein. We can come to a conclusion that RAAS should be responsible mainly for the metabonomics pathway of QGQS granule on antihypertension, and it plays a very important role in protein of profilin-1 inhibition.
Collapse
|
19
|
Hsu PC, Lan RS, Brasky TM, Marian C, Cheema AK, Ressom HW, Loffredo CA, Pickworth WB, Shields PG. Metabolomic profiles of current cigarette smokers. Mol Carcinog 2017; 56:594-606. [PMID: 27341184 PMCID: PMC5646689 DOI: 10.1002/mc.22519] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 05/09/2016] [Accepted: 06/22/2016] [Indexed: 01/07/2023]
Abstract
Smoking-related biomarkers for lung cancer and other diseases are needed to enhance early detection strategies and to provide a science base for tobacco product regulation. An untargeted metabolomics approach by ultra-performance liquid chromatography-quadrupole-time of flight mass spectrometry (UHPLC-Q-TOF MS) totaling 957 assays was used in a novel experimental design where 105 current smokers smoked two cigarettes 1 h apart. Blood was collected immediately before and after each cigarette allowing for within-subject replication. Dynamic changes of the metabolomic profiles from smokers' four blood samples were observed and biomarkers affected by cigarette smoking were identified. Thirty-one metabolites were definitively shown to be affected by acute effect of cigarette smoking, uniquely including menthol-glucuronide, the reduction of glutamate, oleamide, and 13 glycerophospholipids. This first time identification of a menthol metabolite in smokers' blood serves as proof-of-principle for using metabolomics to identify new tobacco-exposure biomarkers, and also provides new opportunities in studying menthol-containing tobacco products in humans. Gender and race differences also were observed. Network analysis revealed 12 molecules involved in cancer, notably inhibition of cAMP. These novel tobacco-related biomarkers provide new insights to the effects of smoking which may be important in carcinogenesis but not previously linked with tobacco-related diseases. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ping-Ching Hsu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210
| | - Renny S. Lan
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210
| | - Theodore M. Brasky
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210
| | - Catalin Marian
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy 300041 Timisoara, Romania
| | - Amrita K. Cheema
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC 20057
| | - Habtom W. Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC 20057
| | | | | | - Peter G. Shields
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
20
|
Hsu PC, Lan RS, Brasky TM, Marian C, Cheema AK, Ressom HW, Loffredo CA, Pickworth WB, Shields PG. Menthol Smokers: Metabolomic Profiling and Smoking Behavior. Cancer Epidemiol Biomarkers Prev 2017; 26:51-60. [PMID: 27628308 PMCID: PMC5386404 DOI: 10.1158/1055-9965.epi-16-0124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 07/26/2016] [Accepted: 08/31/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The use of menthol in cigarettes and marketing is under consideration for regulation by the FDA. However, the effects of menthol on smoking behavior and carcinogen exposure have been inconclusive. We previously reported metabolomic profiling for cigarette smokers, and novelly identified a menthol-glucuronide (MG) as the most significant metabolite directly related to smoking. Here, MG is studied in relation to smoking behavior and metabolomic profiles. METHODS This is a cross-sectional study of 105 smokers who smoked two cigarettes in the laboratory one hour apart. Blood nicotine, MG, and exhaled carbon monoxide (CO) boosts were determined (the difference before and after smoking). Spearman correlation, χ2, and ANCOVA adjusted for gender, race, and cotinine levels for menthol smokers assessed the relationship of MG boost, smoking behavior, and metabolic profiles. Multivariate metabolite characterization using supervised partial least squares-discriminant analysis (PLS-DA) was carried out for the classification of metabolomics profiles. RESULTS MG boost was positively correlated with CO boost, nicotine boost, average puff volume, puff duration, and total smoke exposure. Classification using PLS-DA, MG was the top metabolite discriminating metabolome of menthol versus nonmenthol smokers. Among menthol smokers, 42 metabolites were significantly correlated with MG boost, which linked to cellular functions, such as of cell death, survival, and movement. CONCLUSIONS Plasma MG boost is a new smoking behavior biomarker that may provide novel information over self-reported use of menthol cigarettes by integrating different smoking measures for understanding smoking behavior and harm of menthol cigarettes. IMPACT These results provide insight into the biological effect of menthol smoking. Cancer Epidemiol Biomarkers Prev; 26(1); 51-60. ©2016 AACR.
Collapse
Affiliation(s)
- Ping-Ching Hsu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
- Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Renny S Lan
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Theodore M Brasky
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Catalin Marian
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
- Biochemistry Department, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Amrita K Cheema
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C
| | - Habtom W Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C
| | | | | | - Peter G Shields
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
21
|
Chu Y, Jiang H, Ju J, Li Y, Gong L, Wang X, Yang W, Deng Y. A metabolomic study using HPLC–TOF/MS coupled with ingenuity pathway analysis: Intervention effects of Rhizoma Alismatis on spontaneous hypertensive rats. J Pharm Biomed Anal 2016; 117:446-52. [DOI: 10.1016/j.jpba.2015.09.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 09/20/2015] [Accepted: 09/21/2015] [Indexed: 10/23/2022]
|
22
|
Hypoxia induced HIF-1/HIF-2 activity alters trophoblast transcriptional regulation and promotes invasion. Eur J Cell Biol 2015; 94:589-602. [DOI: 10.1016/j.ejcb.2015.10.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 10/20/2015] [Accepted: 10/21/2015] [Indexed: 01/19/2023] Open
|
23
|
Sun L, Duan J, Jiang Y, Wang L, Huang N, Lin L, Liao Y, Liao W. Metastasis-associated in colon cancer-1 upregulates vascular endothelial growth factor-C/D to promote lymphangiogenesis in human gastric cancer. Cancer Lett 2014; 357:242-253. [PMID: 25444928 DOI: 10.1016/j.canlet.2014.11.035] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/13/2014] [Accepted: 11/14/2014] [Indexed: 12/13/2022]
Abstract
Lymphangiogenesis is actively contributed to lymphatic metastasis in gastric cancer (GC), and vascular endothelial growth factor (VEGF)-C and VEGF-D are key regulators for lymphangiogenesis. Metastasis-associated in colon cancer-1 (MACC1) was reported to be associated with lymph node metastasis in a few clinical studies, while little is known about the role of MACC1 in lymphangiogenesis. Hence, in the present study, we explored the potential role of MACC1 in lymphangiogenesis as well as the underlying mechanisms. By clinical observation, we found a positive relationship between MACC1 and lymphangiogenesis. Besides, similar results were also obtained from in vivo and in vitro studies. With an indirect co-culture system, we got that supernatant from MACC1 overexpressed GC cells accelerated human lymphatic endothelial cells' (HLECs') capacity of tube-like formation through enhancing cell proliferation and migration. Moreover, MACC1 overexpressed xenografts also presented more lymphatic vessels. Furthermore, MACC1 significantly increased the expression of VEGF-C/VEGF-D in GC cells and transplanted tumors, which was subsequently suppressed by c-Met inhibitor. All these data suggested a critical role for MACC1 in lymphatic dissemination of GC, providing evidence that MACC1 upregulated VEGF-C/VEGF-D secretion to promote lymphangiogenesis via c-Met signaling.
Collapse
Affiliation(s)
- Li Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiangman Duan
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yaqi Jiang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Oncology, Subei People's Hospital, Yangzhou University, Yangzhou 225001, China
| | - Lin Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Li Lin
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yulin Liao
- Department of Cardiology and Organ Failure Key Laboratory of Ministry of Education, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
24
|
Schneider G, Sellers ZP, Abdel-Latif A, Morris AJ, Ratajczak MZ. Bioactive lipids, LPC and LPA, are novel prometastatic factors and their tissue levels increase in response to radio/chemotherapy. Mol Cancer Res 2014; 12:1560-73. [PMID: 25033840 DOI: 10.1158/1541-7786.mcr-14-0188] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
UNLABELLED Bioactive lipids are fundamental mediators of a number of critical biologic processes such as inflammation, proliferation, and apoptosis. Rhabdomyosarcoma (RMS) is common in adolescence with histologic subtypes that favor metastasis. However, the factors that influence metastasis are not well appreciated. Here, it is shown that lysophosphatidylcholine (LPC) and its derivative, lysophosphatidic acid (LPA), strongly enhance motility and adhesion of human RMS cells. Importantly, these metastatic-associated phenotypes were observed at physiologic concentrations of these lipids, which naturally occur in biologic fluids. Moreover, the effects of these bioactive lipids were much stronger as compared with known peptide-based prometastatic factors in RMS, such as stromal-derived factor-1 or hepatocyte growth factor/scatter factor. Finally, both LPC and LPA levels were increased in several organs after γ-irradiation or chemotherapy, supporting the hypothesis that radio/chemotherapy induces an unwanted prometastatic environment in these organs. IMPLICATIONS LPC and LPA play a previously underappreciated role in dissemination of RMS and suggest that antimetastatic treatment with specific molecules blocking LPC/LPA activity should be part of standard radio/chemotherapy arsenal.
Collapse
Affiliation(s)
- Gabriela Schneider
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Zachariah Payne Sellers
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Ahmed Abdel-Latif
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, Kentucky
| | - Andrew J Morris
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, Kentucky
| | - Mariusz Z Ratajczak
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky. Department of Physiology Pomeranian Medical University, Szczecin, Poland.
| |
Collapse
|
25
|
Sangani R, Pandya CD, Bhattacharyya MH, Periyasamy-Thandavan S, Chutkan N, Markand S, Hill WD, Hamrick M, Isales C, Fulzele S. Knockdown of SVCT2 impairs in-vitro cell attachment, migration and wound healing in bone marrow stromal cells. Stem Cell Res 2013; 12:354-63. [PMID: 24365600 DOI: 10.1016/j.scr.2013.11.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 10/01/2013] [Accepted: 11/03/2013] [Indexed: 11/28/2022] Open
Abstract
Bone marrow stromal cell (BMSC) adhesion and migration are fundamental to a number of pathophysiologic processes, including fracture and wound healing. Vitamin C is beneficial for bone formation, fracture repair and wound healing. However, the role of the vitamin C transporter in BMSC adhesion, migration and wound healing is not known. In this study, we knocked-down the sodium-dependent vitamin C transporter, SVCT2, the only known transporter of vitamin C in BMSCs, and performed cell adhesion, migration, in-vitro scratch wound healing and F-actin re-arrangement studies. We also investigated the role of oxidative stress on the above processes. Our results demonstrate that both oxidative stress and down-regulation of SVCT2 decreased cell attachment and spreading. A trans-well cell migration assay showed that vitamin C helped in BMSC migration and that knockdown of SVCT2 decreased cell migration. In the in-vitro scratch wound healing studies, we established that oxidative stress dose-dependently impairs wound healing. Furthermore, the supplementation of vitamin C significantly rescued the BMSCs from oxidative stress and increased wound closing. The knockdown of SVCT2 in BMSCs strikingly decreased wound healing, and supplementing with vitamin C failed to rescue cells efficiently. The knockdown of SVCT2 and induction of oxidative stress in cells produced an alteration in cytoskeletal dynamics. Signaling studies showed that oxidative stress phosphorylated members of the MAP kinase family (p38) and that vitamin C inhibited their phosphorylation. Taken together, these results indicate that both the SVCT2 transporter and oxidative stress play a vital role in BMSC attachment, migration and cytoskeletal re-arrangement. BMSC-based cell therapy and modulation of SVCT2 could lead to a novel therapeutic approach that enhances bone remodeling, fracture repair and wound healing in chronic disease conditions.
Collapse
Affiliation(s)
- Rajnikumar Sangani
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | - Chirayu D Pandya
- Department of Psychiatry and Health Behavior, Georgia Regents University, Augusta, GA 30912, USA
| | | | | | - Norman Chutkan
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | - Shanu Markand
- Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA 30912, USA
| | - William D Hill
- Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Mark Hamrick
- Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Carlos Isales
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
26
|
Differential effects of trimetazidine on vascular smooth muscle cell and endothelial cell in response to carotid artery balloon injury in diabetic rats. Int J Cardiol 2013; 167:126-33. [DOI: 10.1016/j.ijcard.2011.12.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 11/12/2011] [Accepted: 12/17/2011] [Indexed: 11/24/2022]
|
27
|
He J, Li Y, Yang X, He X, Zhang H, He J, Zhang L. The feedback regulation of PI3K-miR-19a, and MAPK-miR-23b/27b in endothelial cells under shear stress. Molecules 2012; 18:1-13. [PMID: 23344184 PMCID: PMC6269661 DOI: 10.3390/molecules18010001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 12/12/2012] [Accepted: 12/14/2012] [Indexed: 02/05/2023] Open
Abstract
Mechanical stimulation regulates endothelial cell (EC) functions through the modulation of signaling networks and gene expression. Our recent studies have identified that shear stress regulation of microRNAs (miRs)-19a, 23b and 27b, led to the modulation of EC proliferation. However, the underlying molecular mechanisms by which shear stress regulates these miRs have not been explored. Previous studies showed that shear stress activates multiple signaling pathways, including phosphatidylinositol 3 kinase (PI3K) and mitogen-activated protein kinase (MAPK). In this work we demonstrate that inhibition of the PI3K pathway attenuated the shear-induced miR-19a, and inhibition of the MAPK pathway attenuated miR-23b, 27b. The knockdown of miR-19a using antagomir-19a oligonucleotide (AM19a) decreased the shear-induced PI3K activation; whereas AM-23b, 27b reduced the shear-induced MAPK activation. Furthermore, the overexpression of miR-19a overrode the suppressive effects of PI3K inhibitors on shear-induced PI3K activation; the overexpression of miR-23b, 27b had similar effects on ERK activations, but had little effect on P38 and JNK activation. Our findings suggest a positive feedback loop whereby PI3K and MAPK mediate the shear regulation of miR expression, which in turn modulates the shear-regulated PI3K/MAPK signaling events in ECs.
Collapse
Affiliation(s)
- Jian He
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
- Departments of Bioengineering and Medicine and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-431-8561-9808
| | - Xufang Yang
- Department of Pathophysiology, Mudanjiang Medical College, Mudanjiang 157000, China
| | - Xu He
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Haiying Zhang
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Jin He
- The First Clinical Hospital, Jilin University, Changchun 130021, China
| | - Lihong Zhang
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| |
Collapse
|
28
|
Activation of Ras/MEK/ERK signaling in chronic subdural hematoma outer membranes. Brain Res 2012; 1489:98-103. [DOI: 10.1016/j.brainres.2012.10.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 10/07/2012] [Indexed: 01/01/2023]
|
29
|
Che J, Okigaki M, Takahashi T, Katsume A, Adachi Y, Yamaguchi S, Matsunaga S, Takeda M, Matsui A, Kishita E, Ikeda K, Yamada H, Matsubara H. Endothelial FGF receptor signaling accelerates atherosclerosis. Am J Physiol Heart Circ Physiol 2010; 300:H154-61. [PMID: 20952669 DOI: 10.1152/ajpheart.00075.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Members of the fibroblast growth factor (FGF) family have been clinically applied to the treatment of ischemic diseases because of their strong angiogenic actions. Although tissue ischemia is predominantly caused by atherosclerosis, the roles of endothelial FGF receptors (FGF-Rs) in atherosclerosis remain obscure. We generated endothelial cell (EC)-targeted constitutively active FGF-R2-overexpressing mice, using the Tie2 promoter (Tie2-FGF-R2-Tg), and crossed them with apolipoprotein E (ApoE)-deficient mice (ApoE-KO) to generate Tie2-FGF-R2-Tg/ApoE-deficient mice (Tie2-FGF-R2-Tg/ApoE-KO). After being fed a Western diet for 8 wk, the Tie2-FGF-R2-Tg/ApoE-KO demonstrated 2.0-fold greater atherosclerotic lesion area on the luminal surfaces of the aortas than the ApoE-KO (P < 0.01). The level of p21(Cip1) protein, a cell cycle inhibitor, in the FGF-R2-overexpressing EC was 2.5-fold greater than that in the wild-type (WT) EC at the baseline (P < 0.01). FGF-R2 overexpression in the EC resulted in increased expression of VCAM-1 and ICAM-1, acceleration of apoptosis, and decreased proliferative activity, all of which were normalized by small interfering RNA (siRNA)-mediated knockdown of p21(Cip1) (75% reduction in protein level, P < 0.01). Furthermore, the expression of PDGF-B and Egr-1, a PDGF/p21(Cip1)-inducible transcription factor, in the aortic endothelium of Tie2-FGF-R2-Tg/ApoE-KO was significantly greater than that in ApoE-KO. The proliferation of vascular smooth muscle cells in the aortic media of Tie2-FGF-R2-Tg/ApoE-KO was 2.0-fold higher than that in ApoE-KO (P < 0.01). Thus our study reveals that endothelial FGF-R2 signaling aggravates atherosclerosis by promoting p21(Cip1)-mediated EC dysfunction and cautions against the use of FGF for therapeutic angiogenesis in the setting of atherosclerosis.
Collapse
Affiliation(s)
- Jishan Che
- Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Thrombospondin-1 as a Paradigm for the Development of Antiangiogenic Agents Endowed with Multiple Mechanisms of Action. Pharmaceuticals (Basel) 2010; 3:1241-1278. [PMID: 27713299 PMCID: PMC4034032 DOI: 10.3390/ph3041241] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Revised: 04/20/2010] [Accepted: 04/22/2010] [Indexed: 12/12/2022] Open
Abstract
Uncontrolled neovascularization occurs in several angiogenesis-dependent diseases, including cancer. Neovascularization is tightly controlled by the balance between angiogenic growth factors and antiangiogenic agents. The various natural angiogenesis inhibitors identified so far affect neovascularization by different mechanisms of action. Thrombospondin-1 (TSP-1) is a matricellular modular glycoprotein that acts as a powerful endogenous inhibitor of angiogenesis. It acts both indirectly, by sequestering angiogenic growth factors and effectors in the extracellular environment, and directly, by inducing an antiangiogenic program in endothelial cells following engagement of specific receptors including CD36, CD47, integrins and proteoglycans (all involved in angiogenesis ). In view of its central, multifaceted role in angiogenesis, TSP-1 has served as a source of antiangiogenic tools, including TSP-1 fragments, synthetic peptides and peptidomimetics, gene therapy strategies, and agents that up-regulate TSP-1 expression. This review discusses TSP-1-based inhibitors of angiogenesis, their mechanisms of action and therapeutic potential, drawing our experience with angiogenic growth factor-interacting TSP-1 peptides, and the possibility of exploiting them to design novel antiangiogenic agents.
Collapse
|
31
|
Kimura T, Tomura H, Sato K, Ito M, Matsuoka I, Im DS, Kuwabara A, Mogi C, Itoh H, Kurose H, Murakami M, Okajima F. Mechanism and role of high density lipoprotein-induced activation of AMP-activated protein kinase in endothelial cells. J Biol Chem 2009; 285:4387-97. [PMID: 20018878 DOI: 10.1074/jbc.m109.043869] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The upstream signaling pathway leading to the activation of AMP-activated protein kinase (AMPK) by high density lipoprotein (HDL) and the role of AMPK in HDL-induced antiatherogenic actions were investigated. Experiments using genetic and pharmacological tools showed that HDL-induced activation of AMPK is dependent on both sphingosine 1-phosphate receptors and scavenger receptor class B type I through calcium/calmodulin-dependent protein kinase kinase and, for scavenger receptor class B type I system, additionally serine-threonine kinase LKB1 in human umbilical vein endothelial cells. HDL-induced activation of Akt and endothelial NO synthase, stimulation of migration, and inhibition of monocyte adhesion and adhesion molecule expression were dependent on AMPK activation. The inhibitory role of AMPK in the adhesion molecule expression and monocyte adhesion on endothelium of mouse aorta was confirmed in vivo and ex vivo. On the other hand, stimulation of ERK and proliferation were hardly affected by AMPK knockdown but completely inhibited by an N17Ras, whereas the dominant-negative Ras was ineffective for AMPK activation. In conclusion, dual HDL receptor systems differentially regulate AMPK activity through calcium/calmodulin-dependent protein kinase kinase and/or LKB1. Several HDL-induced antiatherogenic actions are regulated by AMPK, but proliferation-related actions are regulated by Ras rather than AMPK.
Collapse
Affiliation(s)
- Takao Kimura
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ali M, Madjid M. Lipoprotein-associated phospholipase A2: a cardiovascular risk predictor and a potential therapeutic target. Future Cardiol 2009; 5:159-73. [DOI: 10.2217/14796678.5.2.159] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Lipoprotein-associated phospholipase A2 (Lp-PLA2), present in the circulation and in atherosclerotic plaque, is an inflammatory marker with potential use as a predictor of cardiovascular risk and as a therapeutic target. Although Lp-PLA2 is associated with both LDL and HDL, it is important to determine whether Lp-PLA2 has a predominantly pro- or anti-atherogenic effect. Increasing evidence suggests a proatherogenic role for Lp-PLA2. ©iEpidemiologic and clinical evidence suggests Lp-PLA2 is an independent predictor of risk and may be superior to other inflammatory markers owing to its specificity and minimal biovariation. Lp-PLA2 inhibitors currently being investigated in clinical trials are promising novel anti-inflammatory agents with a specificity for the vascular bed and a potential for decreasing plaque vulnerability.
Collapse
Affiliation(s)
- Muzammil Ali
- Texas Heart Institute, 6770 Bertner Ave, MC 2-255, Houston, TX 77030, USA
| | - Mohammad Madjid
- Texas Heart Institute at St Luke’s Episcopal Hospital, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
33
|
Nishikawa T, Nakagami H, Maeda A, Morishita R, Miyazaki N, Ogawa T, Tabata Y, Kikuchi Y, Hayashi H, Tatsu Y, Yumoto N, Tamai K, Tomono K, Kaneda Y. Development of a novel antimicrobial peptide, AG-30, with angiogenic properties. J Cell Mol Med 2008; 13:535-46. [PMID: 18410525 PMCID: PMC3822513 DOI: 10.1111/j.1582-4934.2008.00341.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The utility of various synthetic peptides has been investigated in clinical trials of the treatment of cancers, infectious diseases and endocrine diseases. In the process of functional gene screening with in silico analysis for molecules with angiogenic properties, we generated a small peptide, angiogenic peptide (AG)-30, that possesses both antimicrobial and pro-inflammatory activities. AG-30 has an α-helix structure with a number of hydrophobic or net positively charged amino acids and a propensity to fold into amphipathic structures. Indeed, AG-30 exhibited antimicrobial activity against various bacteria, induced vascular endothelial cell growth and tube formation in a dose-dependent manner and increased neovascularization in a Matrigel plug assay. As a result, AG-30 up-regulated expression of angiogenesis-related cytokines and growth factors for up to 72 hrs in human aortic endothelial cells. To further evaluate the angiogenic effect of AG-30 in vivo, we developed a slow-release AG-30 system utilizing biodegradable gelatin microspheres. In the ischaemic mouse hind limb, slow-release AG-30 treatment results in an increase in angiogenic score, an increase in blood flow (as demonstrated by laser Doppler imaging) and an increase in capillary density (as demonstrated by immunostaining with anti-CD31 antibody). These data suggest that the novel peptide, AG-30, may have therapeutic potential for ischaemic diseases.
Collapse
Affiliation(s)
- Tomoyuki Nishikawa
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Saito Y, Nakagami H, Kurooka M, Takami Y, Kikuchi Y, Hayashi H, Nishikawa T, Tamai K, Morishita R, Azuma N, Sasajima T, Kaneda Y. Cold shock domain protein A represses angiogenesis and lymphangiogenesis via inhibition of serum response element. Oncogene 2007; 27:1821-33. [DOI: 10.1038/sj.onc.1210824] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
35
|
Zhu S, Korzh V, Gong Z, Low BC. RhoA prevents apoptosis during zebrafish embryogenesis through activation of Mek/Erk pathway. Oncogene 2007; 27:1580-9. [PMID: 17873909 DOI: 10.1038/sj.onc.1210790] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
RhoA small GTPase, as a key regulator for actin cytoskeletal rearrangement, plays pivotal roles during morphogenesis, cytokinesis, phagocytosis and cell migration, but little is known about its signaling mechanism that controls cell survival in vivo. Using zebrafish as a model, we show that non-overlapping antisense morpholinos that block either translation or splicing of rhoA lead to extensive apoptosis during embryogenesis, resulting in overall reduction of body size and body length. These defects are associated with reduced activation of growth-promoting Erk and decreased expression of anti-apoptotic bcl-2. Moreover, ectopic expression of rhoA, Mek or BCL-2 mRNA rescues such phenotypes. Consistently, combined suppression of RhoA and Mek/Erk or Bcl-2 pathways by sub-optimal dose of rhoA morpholino and pharmacological inhibitors for either Mek (U0126) or Bcl-2 (HA 14-1) can induce developmental abnormalities and enhanced apoptosis, similar to those caused by effective RhoA knockdown. Furthermore, U0126 abrogates the rescue by RhoA and MEK but not BCL-2. In contrast, HA 14-1 effectively abolishes all functional rescues by RhoA, MEK or BCL-2, supporting that RhoA prevents apoptosis by activation of Mek/Erk pathway and requiring Bcl-2. These findings reveal an important genetic and functional relationship between RhoA with Mek/Erk and Bcl-2 for cell survival control during embryogenesis.
Collapse
Affiliation(s)
- S Zhu
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore
| | | | | | | |
Collapse
|
36
|
Tang J, Hu G, Hanai JI, Yadlapalli G, Lin Y, Zhang B, Galloway J, Bahary N, Sinha S, Thisse B, Thisse C, Jin JP, Zon LI, Sukhatme VP. A critical role for calponin 2 in vascular development. J Biol Chem 2005; 281:6664-72. [PMID: 16317011 DOI: 10.1074/jbc.m506991200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Calponin 2 (h2 calponin, CNN2) is an actin-binding protein implicated in cytoskeletal organization. We have found that the expression of calponin 2 is relatively restricted to vasculature from 16 to 30 h post-fertilization during zebrafish (Danio rerio) development. Forty-eight hours after injecting antisense morpholino oligos against calponin 2 into embryos at the 1-4-cell stage, zebrafish demonstrated various cardiovascular defects, including sluggish axial and head circulation, absence of circulation in intersegmental vessels and in the dorsal longitudinal anastomotic vessel, enlarged cerebral ventricles, and pericardial edema, in addition to an excess bending, spiraling tail and twisting of the caudal fin. Knockdown of calponin 2 in the Tg(fli1:EGFP)(y1) zebrafish line (in which a fli1 promoter drives vascular-specific enhanced green fluorescent protein expression) indicated that diminished calponin 2 expression blocked the proper migration of endothelial cells during formation of intersegmental vessels. In vitro studies showed that basic fibroblast growth factor-induced human umbilical vein endothelial cell migration was down-regulated by knockdown of calponin 2 expression using an antisense adenovirus, and overexpression of calponin 2 enhanced migration and hastened wound healing. These events were correlated with activation of mitogen-activated protein kinase; moreover, inhibition of this pathway blocked the promigratory effect of calponin 2. Collectively, these data suggest that calponin 2 plays an important role in the migration of endothelial cells both in vivo and in vitro and that its expression is critical for proper vascular development.
Collapse
Affiliation(s)
- Jian Tang
- Renal Division and Center for Study of the Tumor Microenvironment, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Matsusaka S, Wakabayashi I. 5-Hydroxytryptamine as a potent migration enhancer of human aortic endothelial cells. FEBS Lett 2005; 579:6721-5. [PMID: 16310780 DOI: 10.1016/j.febslet.2005.10.068] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2005] [Revised: 10/24/2005] [Accepted: 10/27/2005] [Indexed: 01/09/2023]
Abstract
The purpose of the present study was to investigate whether 5-hydroxytryptamine (5-HT, serotonin) affects migration of vascular endothelial cells. 5-HT significantly enhanced migration of human aortic endothelial cells (HAECs), and this enhancement was completely inhibited by GR 55562, a 5-HT1 receptor antagonist, and fluoxetine, a 5-HT transporter inhibitor, but was not affected by ketanserin, a 5-HT2 receptor antagonist. 5-HT stimulation increased RhoA and ERK activity of HAECs, and inhibitors of RhoA (Y-27632 and H-1152) and inhibitors of MEK (U0126 and PD98059) abolished the 5-HT-induced increase in migration velocity. Inhibition of Rho kinase by Y-27632 blocked stress fiber formation and rear release of HAECs. Thus, 5-HT has a potent enhancing action on migration of HAECs through activating the RhoA and ERK pathways following 5-HT1 receptor stimulation.
Collapse
Affiliation(s)
- Satoshi Matsusaka
- Department of Hygiene and Preventive Medicine, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan
| | | |
Collapse
|
38
|
Macphee C, Benson GM, Shi Y, Zalewski A. Lipoprotein-associated phospholipase A2: a novel marker of cardiovascular risk and potential therapeutic target. Expert Opin Investig Drugs 2005; 14:671-9. [PMID: 16004595 DOI: 10.1517/13543784.14.6.671] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Although the clinical benefit of statins is well established, these agents reduce the risk of cardiovascular events by only 20 - 40%, and the residual risk for high-risk patients is considerable. The recognition of atherosclerosis as an inflammatory disease has opened the door to numerous complementary therapeutic approaches to further reduce risk and the overall burden of cardiovascular disease. Lipoprotein-associated phospholipase A(2) (Lp-PLA(2)) is a novel inflammatory marker of cardiovascular risk that is being evaluated as a potential therapeutic target. The biological function of this enzyme in atherosclerosis has been controversial but recent evidence supports its pro-atherogenic role. The enzyme is predominantly bound to low-density lipoprotein cholesterol particles in humans, and its activity produces bioactive lipid mediators that promote inflammatory processes present at every stage of atherogenesis, from atheroma initiation to plaque destabilisation and rupture. Initial clinical studies suggest that the inhibitors of Lp-PLA(2) can block enzyme activity in plasma and within atherosclerotic plaques. However, more studies are needed to determine the potential clinical benefits of inhibiting Lp-PLA(2).
Collapse
Affiliation(s)
- Colin Macphee
- GlaxoSmithKline, Vascular Biology and Thrombosis, 709 Swedeland Road, King of Prussia, PA 19406, USA.
| | | | | | | |
Collapse
|
39
|
Huang TY, Chen HI, Liu CY, Jen CJ. Lysophosphatidylcholine alters vascular tone in rat aorta by suppressing endothelial [Ca(2+)](I) signaling. J Biomed Sci 2002; 9:327-33. [PMID: 12145530 DOI: 10.1007/bf02256588] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The detailed mechanism of how lysophosphatidylcholine (LPC) suppresses endothelium-dependent vasodilatation is unclear at present. We investigated the effects of LPC on endothelial intracellular calcium (EC [Ca(2+)](i)) signaling and vascular tone simultaneously using a new technique we developed. Fura-2-labeled rat aortic specimens were mounted in a tissue flow chamber and precontracted with phenylephrine (5 x 10(-8) M). Under either basal or agonist-stimulated conditions, the EC [Ca(2+)](i) level was calculated from fura 2 fluorescence ratio images, and the vascular tone was estimated by measuring the relative displacement of the fluorescence images. Although both acetylcholine (ACh)-induced EC [Ca(2+)](i) elevation and the concomitant vasorelaxation were partially suppressed in specimens pretreated with LPC (20 microM), the quantitative relationship between EC [Ca(2+)](i) elevation and the corresponding vasorelaxation was unaffected. A high concentration of LPC (40 microM) completely eliminated ACh-evoked [Ca(2+)](i) elevation and vasodilatation. It has been reported that exposing vascular tissue to a calcium-free buffer causes a reduction in the EC [Ca(2+)](i) level and the accompanying vasoconstriction. Pretreatment with 20 microM LPC reduced the basal EC [Ca(2+)](i) level and abolished the calcium-free solution-induced EC [Ca(2+)](i) reduction and vasoconstriction. We conclude that LPC impairs endothelium-dependent vasorelaxation mainly by reducing the basal EC [Ca(2+)](i) level and suppressing agonist-evoked EC [Ca(2+)](i) signaling.
Collapse
Affiliation(s)
- Tung-Yi Huang
- Department of Physiology and Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | | | | | | |
Collapse
|
40
|
Goetze S, Eilers F, Bungenstock A, Kintscher U, Stawowy P, Blaschke F, Graf K, Law RE, Fleck E, Gräfe M. PPAR activators inhibit endothelial cell migration by targeting Akt. Biochem Biophys Res Commun 2002; 293:1431-7. [PMID: 12054675 DOI: 10.1016/s0006-291x(02)00385-6] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) regulate lipid and glucose metabolism and exert several vascular effects that may provide a dual benefit of these receptors on metabolic disorders and atherosclerotic vascular disease. Endothelial cell migration is a key event in the pathogenesis of atherosclerosis. We therefore investigated the effects of lipid-lowering PPARalpha-activators (fenofibrate, WY14643) and antidiabetic PPARgamma-activators (troglitazone, ciglitazone) on this endothelial cell function. Both PPARalpha- and PPARgamma-activators significantly inhibited VEGF-induced migration of human umbilical vein endothelial cells (EC) in a concentration-dependent manner. Chemotactic signaling in EC is known to require activation of two signaling pathways: the phosphatidylinositol-3-kinase (PI3K)-->Akt- and the ERK1/2 mitogen-activated protein kinase (ERK MAPK) pathway. Using the pharmacological PI3K-inhibitor wortmannin and the ERK MAPK-pathway inhibitor PD98059, we observed a complete inhibition of VEGF-induced EC migration. VEGF-induced Akt phosphorylation was significantly inhibited by both PPARalpha- and gamma-activators. In contrast, VEGF-stimulated ERK MAPK-activation was not affected by any of the PPAR-activators, indicating that they inhibit migration either downstream of ERK MAPK or independent from this pathway. These results provide first evidence for the antimigratory effects of PPAR-activators in EC. By inhibiting EC migration PPAR-activators may protect the vasculature from pathological alterations associated with metabolic disorders.
Collapse
Affiliation(s)
- Stephan Goetze
- Department of Medicine/Cardiology, German Heart Institute Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Hwang KC, Lee KH, Jang Y, Yun YP, Chung KH. Epigallocatechin-3-gallate inhibits basic fibroblast growth factor-induced intracellular signaling transduction pathway in rat aortic smooth muscle cells. J Cardiovasc Pharmacol 2002; 39:271-7. [PMID: 11791013 DOI: 10.1097/00005344-200202000-00014] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Daily green tea drinking showed preventive effects on the progression of atherosclerosis. Although epigallocatechin-3-gallate [EGCG] has anti-proliferative effects on various cells, relatively little is known about the molecular mechanisms of the anti-proliferative effects of EGCG. To determine whether the transduction signals and protooncogene expression were affected by EGCG, this study investigated the molecular mechanism of the anti-proliferative effects in basic fibroblast growth factor (bFGF)-stimulated rat aortic smooth muscle cells (RAoSMCs). EGCG inhibited the proliferative response stimulated by 10% fetal bovine serum dose dependently in RAoSMCs (median inhibitory concentration [IC50]: 28.4 x 10(-6) M ). EGCG also inhibited the migration of bFGF-stimulated RAoSMCs in a dose-dependent manner, showing that 21.8 x 10(-6) M of EGCG significantly inhibited the migration by 75 +/- 5% in comparison with bFGF-stimulated migration. In RAoSMCs, EGCG dramatically inhibited Ras activation and c-jun N-terminal kinase (JNK) activity without affecting protein kinase C expression. Induction of c-jun mRNA stimulated by bFGF was significantly reduced dose dependently up to 87.3 x 10(-6) M of EGCG. These results indicate that the anti-proliferative effect of EGCG on RAoSMCs is partly Ras/JNK mediated, independent of protein kinase C, and is attributable to the downregulation of c-jun expression.
Collapse
Affiliation(s)
- Ki-Chul Hwang
- Cardiovascular Research Institute, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul
| | | | | | | | | |
Collapse
|
42
|
Kutz SM, Hordines J, McKeown-Longo PJ, Higgins PJ. TGF-β1-induced PAI-1 gene expression requires MEK activity and cell-to-substrate adhesion. J Cell Sci 2001; 114:3905-14. [PMID: 11719557 DOI: 10.1242/jcs.114.21.3905] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The type-1 inhibitor of plasminogen activator (PAI-1) is an important physiological regulator of extracellular matrix (ECM) homeostasis and cell motility. Various growth factors mediate temporal changes in the expression and/or focalization of PAI-1 and its protease target PAs, thereby influencing cell migration by barrier proteolysis and/or ECM adhesion modulation. TGF-β1, in particular, is an effective inducer of matrix deposition/turnover, cell locomotion and PAI-1 expression. Therefore, the relationship between motility and PAI-1 induction was assessed in TGF-β1-sensitive T2 renal epithelial cells. PAI-1 synthesis and its matrix deposition in response to TGF-β1 correlated with a significant increase in cell motility. PAI-1 expression was an important aspect in cellular movement as PAI-1-deficient cells had significantly impaired basal locomotion and were unresponsive to TGF-β1. However, the induced migratory response to this growth factor was complex. TGF-β1 concentrations of 1-2 ng/ml were significantly promigratory, whereas lower levels (0.2-0.6 ng/ml) were ineffective and final concentrations ≥5 ng/ml inhibited T2 cell motility. This same growth factor range progressively increased PAI-1 transcript levels in T2 cells consistent with a bifunctional role for PAI-1 in cell migration. TGF-β1 induced PAI-1 mRNA transcripts in quiescent T2 cells via an immediate-early response mechanism. Full TGF-β1-stimulated expression required tyrosine kinase activity and involved MAPK/ERK kinase (MEK). MEK appeared to be a major mediator of TGF-β1-dependent PAI-1 expression and T2 cell motility since PD98059 effectively attenuated both TGF-β1-induced ERK1/2 activation and PAI-1 transcription as well as basal and growth factor-stimulated planar migration. Since MEK activation in response to growth factors is adhesion-dependent, it was important to determine whether cellular adhesive state influenced TGF-β1-mediated PAI-1 expression in the T2 cell system. Cells maintained in suspension culture (i.e., over agarose underlays) in growth factor-free medium or treated with TGF-β1 in suspension expressed relatively low levels of PAI-1 transcripts compared with the significant induction of PAI-1 mRNA evident in T2 cells upon stimulation with TGF-β1 during adhesion to a fibronectin-coated substrate. Attachment to fibronectin alone (i.e., in the absence of added growth factor) was sufficient to initiate PAI-1 transcription, albeit at levels considerably lower than that induced by the combination of cell adhesion in the presence of TGF-β1. T2 cells allowed to attach to vitronectin-coated surfaces also expressed PAI-1 transcripts but to a significantly reduced extent relative to cells adherent to fibronectin. Moreover, newly vitronectin-attached cells did not exhibit a PAI-1 inductive response to TGF-β1, at least during the short 2 hour period of combined treatment. PAI-1 mRNA synthesis in response to substrate attachment, like TGF-β1-mediated induction in adherent cultures, also required MEK activity as fibronectin-stimulated PAI-1 expression was effectively attenuated by the MEK inhibitor PD98059. These data indicate that cellular adhesive state modulates TGF-β1 signaling to particular target genes (i.e., PAI-1) and that MEK is a critical mediator of the PAI-1+/promigratory phenotype switch induced by TGF-β1 in T2 cells.
Collapse
Affiliation(s)
- S M Kutz
- Center for Cell Biology & Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | | | | | | |
Collapse
|
43
|
Brill A, Franitza S, Lider O, Hershkoviz R. Regulation of T-cell interaction with fibronectin by transforming growth factor-beta is associated with altered Pyk2 phosphorylation. Immunology 2001; 104:149-56. [PMID: 11683954 PMCID: PMC1783298 DOI: 10.1046/j.1365-2567.2001.01283.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although the involvement of transforming growth factor-beta (TGF-beta) in inflammatory reactions has been extensively studied, its mode of action in the context of the extracellular matrix (ECM) is still not fully understood. We undertook this study in an attempt to reveal the putative roles of TGF-beta in T-cell adhesion and migration. We found that a 60-min treatment of T cells with TGF-beta regulates T-cell adhesion to fibronectin (FN), a prototype cell adhesion protein of the ECM, depending on the presence of other activators. At 5 pg/ml to 1 ng/ml, TGF-beta alone induced T-cell adhesion to FN in an integrin alpha4/beta1- and integrin alpha5/beta1-dependent manner. TGF-beta also attenuated T-cell migration on the stromal cell-derived factor (SDF)-1alpha gradients. These effects of TGF-beta were not accompanied by alteration in the expression of very-late activation antigen type 4 (VLA-4) and VLA-5, nor were they mediated by the cyclo-oxygenase pathway. The cellular mechanism underlying the adhesion-regulating activities of TGF-beta involves adhesion-associated cytoskeletal elements. TGF-beta induced the phosphorylation of focal adhesion kinase Pyk2, but not extracellular signal-regulated kinase (ERK), and this effect was markedly increased in the presence of immobilized FN, suggesting a collaborative role for FN-specific integrins. Indeed, TGF-beta-induced Pyk2 phosphorylation was inhibited by monoclonal antibodies against VLA-4, VLA-5 and CD29. Thus, TGF-beta, which may appear at extravascular sites during inflammation, affects the adhesion of T cells to ECM glycoproteins and their migration by its ability to differentially induce or inhibit the phosphorylation of Pyk2.
Collapse
Affiliation(s)
- Alexander Brill
- Department of Immunology, The Weizmann Institute of ScienceRehovot, Israel
| | - Susanne Franitza
- Department of Immunology, The Weizmann Institute of ScienceRehovot, Israel
| | - Ofer Lider
- Department of Immunology, The Weizmann Institute of ScienceRehovot, Israel
| | | |
Collapse
|
44
|
Rikitake Y, Kawashima S, Takahashi T, Ueyama T, Ishido S, Inoue N, Hirata K, Yokoyama M. Regulation of tyrosine phosphorylation of PYK2 in vascular endothelial cells by lysophosphatidylcholine. Am J Physiol Heart Circ Physiol 2001; 281:H266-74. [PMID: 11406493 DOI: 10.1152/ajpheart.2001.281.1.h266] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Lysophosphatidylcholine (LPC), a component of oxidized low-density lipoprotein, exerts various biological effects on vascular endothelial cells. However, the intracellular signaling of LPC is poorly understood. In this study, we investigated the involvement of proline-rich tyrosine kinase (PYK2) in LPC signaling in cultured bovine aortic endothelial cells by immunoprecipitation and Western blotting assays. Treatment of cells with LPC promoted a rapid increase in tyrosine phosphorylation of PYK2. LPC-stimulated PYK2 phosphorylation was inhibited by calcium chelators, 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester, EGTA, protein kinase C (PKC) inhibitor, GF-109203X, or PKC depletion by phorbol esters. PYK2 phosphorylation was inhibited by treatment with cytochalasin D but with neither botulinum C3 transferase nor overexpression of a dominant negative mutant of Rho A. LPC stimulated the association of Shc with PYK2, Shc tyrosine phosphorylation, and Grb2 binding to Shc and induced Ras activation. These results provide evidence that 1) LPC tyrosine phosphorylates PYK2 by calcium- and PKC-dependent mechanisms, 2) the intact cytoskeleton is required for LPC-stimulated PYK2 phosphorylation, and 3) LPC-activated Ras via the PYK2/Shc/Grb2 signaling.
Collapse
Affiliation(s)
- Y Rikitake
- First Department of Internal Medicine, Kobe University School of Medicine, Kobe 650-0017, Japan
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Macphee CH. Lipoprotein-associated phospholipase A2: a potential new risk factor for coronary artery disease and a therapeutic target. Curr Opin Pharmacol 2001; 1:121-5. [PMID: 11714085 DOI: 10.1016/s1471-4892(01)00024-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The recognition that atherosclerosis represents an inflammatory disease has begun to shift interest towards novel therapies that could specifically target the underlying inflammatory component of atherogenesis. Like low-density lipoprotein, an ideal new drug target would be a modifiable plasma risk factor that not only reflects the ongoing inflammatory process but also actively promotes it. Lipoprotein-associated phospholipase A2, also known as platelet-activating factor acetylhydrolase, is a new risk factor that may have the potential to fulfil these requirements.
Collapse
Affiliation(s)
- C H Macphee
- Department of Vascular Biology, SmithKline Beecham Pharmaceuticals, Harlow, Essex, UK.
| |
Collapse
|
46
|
Zubilewicz A, Hecquet C, Jeanny JC, Soubrane G, Courtois Y, Mascarelli F. Two distinct signalling pathways are involved in FGF2-stimulated proliferation of choriocapillary endothelial cells: a comparative study with VEGF. Oncogene 2001; 20:1403-13. [PMID: 11313884 DOI: 10.1038/sj.onc.1204231] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2000] [Revised: 12/21/2000] [Accepted: 01/03/2001] [Indexed: 01/01/2023]
Abstract
In the retina, angiogenesis is an important component of normal physiological events such as embryonic vascular development. It is also involved in pathological processes including diabetic retinopathies and age-related macular degeneration, and tumour growth such as choroidal melanoma. Fibroblast growth factor (FGF) 2 and vascular endothelial cell growth factor (VEGF) are the two major angiogenic factors in the retina. We investigated the mechanism of proliferation and the regulation of the mitogenic properties of FGF2 and VEGF in cultures of chorocapillary endothelial cells (CEC). FGF2 is a strong mitogen for CEC and induced a 2.5-fold increase in cell proliferation after 4 days in culture in the absence of serum. In contrast, VEGF is a poor mitogen for CEC. FGF2, but not VEGF induces a large activation of MEK1, ERK1/2 and P90(RSK) during CEC proliferation. Pharmacological inhibition of Ras processing, and of MEK1 and ERK1/2 activation reduced only by 50% FGF2-induced cell proliferation, suggesting that there is another signalling pathway for CEC proliferation. Pharmacological inhibition of the PI 3-Kinase also inhibits by half FGF2-induced CEC proliferation. FGF2 stimulates the activation of the PI 3-K, P70(S6K) and Akt. Inhibition of both ERK1/2 and PI 3-K activities suppressed FGF2-induced CEC proliferation, demonstrating that CEC proliferation requires both ERKs and PI 3-K pathways. These data on the molecular mechanism and signalling may have important implications for providing more selective methods for anti-angiogenic and anti-tumoural therapy.
Collapse
Affiliation(s)
- A Zubilewicz
- Medical School in Lublin, Department of Ophthalmology, ul. Chmielna 1, 20-079, Lublin, Poland
| | | | | | | | | | | |
Collapse
|
47
|
Rikitake Y, Hirata K, Kawashima S, Takeuchi S, Shimokawa Y, Kojima Y, Inoue N, Yokoyama M. Signaling mechanism underlying COX-2 induction by lysophosphatidylcholine. Biochem Biophys Res Commun 2001; 281:1291-7. [PMID: 11243876 DOI: 10.1006/bbrc.2001.4510] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Lysophosphatidylcholine, a component of oxidized low density lipoprotein, is critical for pathological conditions including atherosclerosis. However, the signaling mechanism of lysophosphatidylcholine remains poorly understood. Here we reported that lysophosphatidylcholine induces phosphorylation of p38 and the transcription factors, CREB and ATF-1 with concomitant up-regulation of cyclooxygenase-2 expression in cultured vascular endothelial cells. Lysophosphatidylcholine induced p38 phosphorylation in a time- and concentration-dependent manner partly via pathway depending on protein tyrosine kinase. Both lysophosphatidylcholine-stimulated phosphorylation of CREB and ATF-1 and lysophosphatidylcholine-increased expression of cyclooxygenase-2 mRNA and protein were effectively inhibited by a combination of SB203580 and PD98059, specific inhibitors of p38 and MEK1, respectively, as well as Ro31-8220 and H89, potent inhibitors of MSK1. These results suggest that both p38 and ERK may function as upstream signaling pathways capable of activating CREB and ATF-1 with subsequent induction of cyclooxygenase-2 expression by lysophosphatidylcholine.
Collapse
Affiliation(s)
- Y Rikitake
- First Department of Internal Medicine, Kobe University School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Cieslik K, Abrams CS, Wu KK. Up-regulation of endothelial nitric-oxide synthase promoter by the phosphatidylinositol 3-kinase gamma /Janus kinase 2/MEK-1-dependent pathway. J Biol Chem 2001; 276:1211-9. [PMID: 11042169 DOI: 10.1074/jbc.m005305200] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Our recent study indicates that lysophosphatidylcholine (LPC) enhances Sp1 binding and Sp1-dependent endothelial nitric oxide synthase (eNOS) promoter activity via the mitogen-activated protein kinase/extracellular signal-regulated kinase kinase 1 (MEK-1) signaling pathway (Cieslik, K., Lee, C.-M., Tang, J.-L., and Wu, K. K. (1999) J. Biol. Chem. 274, 34669-34675). To identify upstream signaling molecules, we transfected human endothelial cells with dominant negative and active mutants of Ras and evaluated their effects on eNOS promoter activity. Neither mutant altered the basal or LPC-induced eNOS promoter function. By contrast, a dominant negative mutant of phosphatidylinositol 3-kinase gamma (PI-3Kgamma) blocked the promoter activity induced by LPC. Wortmannin and LY 294002 had a similar effect. AG-490, a selective inhibitor of Janus kinase 2 (Jak2), also reduced the LPC-induced Sp1 binding and eNOS promoter activity to the basal level. LPC induced Jak2 phosphorylation, which was abolished by LY 294002 and the dominant negative mutant of PI-3Kgamma. LY 294002 and AG-490 abrogated MEK-1 phosphorylation induced by LPC but had no effect on Raf-1. These results indicate that PI-3Kgamma and Jak2 are essential for LPC-induced eNOS promoter activity. This signaling pathway was sensitive to pertussis toxin, suggesting the involvement of a G(i) protein in PI-3Kgamma activation. These results indicate that LPC enhances Sp1-dependent eNOS promoter activity by a pertussis toxin-sensitive, Ras-independent novel pathway, PI-3Kgamma/Jak2/MEK-1/ERK1/2.
Collapse
Affiliation(s)
- K Cieslik
- Vascular Biology Research Center and Division of Hematology, University of Texas Medical School, Houston, Texas 77030, USA
| | | | | |
Collapse
|