1
|
Yegambaram M, Kumar S, Wu X, Lu Q, Sun X, Garcia Flores A, Meadows ML, Barman S, Fulton D, Wang T, Fineman JR, Black SM. Endothelin-1 acutely increases nitric oxide production via the calcineurin mediated dephosphorylation of Caveolin-1. Nitric Oxide 2023; 140-141:50-57. [PMID: 37659679 DOI: 10.1016/j.niox.2023.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/22/2023] [Accepted: 08/30/2023] [Indexed: 09/04/2023]
Abstract
Endothelin (ET)-1 is an endothelial-derived peptide that exerts biphasic effects on nitric oxide (NO) levels in endothelial cells such that acute exposure stimulates-while sustained exposure attenuates-NO production. Although the mechanism involved in the decrease in NO generation has been identified but the signaling involved in the acute increase in NO is still unresolved. This was the focus of this study. Our data indicate that exposing pulmonary arterial endothelial cells (PAEC) to ET-1 led to an increase in NO for up to 30min after which levels declined. These effects were attenuated by ET receptor antagonists. The increase in NO correlated with significant increases in pp60Src activity and increases in eNOS phosphorylation at Tyr83 and Ser1177. The ET-1 mediated increase in phosphorylation and NO generation were attenuated by the over-expression of a pp60Src dominant negative mutant. The increase in pp60Src activity correlated with a reduction in the interaction of Caveolin-1 with pp60Src and the calcineurin-mediated dephosphorylation of caveolin-1 at three previously unidentified sites: Thr91, Thr93, and Thr95. The calcineurin inhibitor, Tacrolimus, attenuated the acute increase in pp60Src activity induced by ET-1 and a calcineurin siRNA attenuated the ET-1 mediated increase in eNOS phosphorylation at Tyr83 and Ser1177 as well as the increase in NO. By using a Caveolin-1 celluSpot peptide array, we identified a peptide targeting a sequence located between aa 41-56 as the pp60Src binding region. This peptide fused to the TAT sequence was found to decrease caveolin-pp60Src interaction, increased pp60Src activity, increased eNOS pSer1177 and NO levels in PAEC and induce vasodilation in isolated aortic rings in wildtype but not eNOS knockout mice. Together, our data identify a novel mechanism by which ET-1 acutely increases NO via a calcineurin-mediated dephosphorylation of caveolin-1 and the subsequent stimulation of pp60Src activity, leading to increases in phosphorylation of eNOS at Tyr83 and Ser1177.
Collapse
Affiliation(s)
- Manivannan Yegambaram
- Center of Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA
| | - Sanjiv Kumar
- Department of Medicine, Augusta University, Augusta, GA, USA; Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Xiaomin Wu
- Department of Medicine, University of Arizona, Tucson, AZ, 33174, USA
| | - Qing Lu
- Center of Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA
| | - Xutong Sun
- Center of Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA
| | - Alejandro Garcia Flores
- Center of Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA
| | | | - Scott Barman
- Department of Pharmacology, Augusta University, Augusta, GA, USA
| | - David Fulton
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Department of Pharmacology, Augusta University, Augusta, GA, USA
| | - Ting Wang
- Center of Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Stephen M Black
- Center of Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33174, USA.
| |
Collapse
|
2
|
Chen X, Zou Z, Wang Q, Gao W, Zeng S, Ye S, Xu P, Huang M, Li K, Chen J, Zhong Z, Zhang Q, Hao B, Liu Q. Inhibition of NOS1 promotes the interferon response of melanoma cells. J Transl Med 2022; 20:205. [PMID: 35538490 PMCID: PMC9092760 DOI: 10.1186/s12967-022-03403-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/22/2022] [Indexed: 02/07/2023] Open
Abstract
Background NOS1 expression predicts poor prognosis in patients with melanoma. However, the molecular function of NOS1 in the type I IFN response and immune escape of melanoma is still unknown. Methods The CRISPR/Cas9 system was used to generate NOS1-knockout melanoma cells and the biological characteristics of NOS1-knockout cells were evaluated by MTT assay, clonogenic assay, EdU assay, and flow cytometric assay. The effect on tumor growth was tested in BALB/c-nu and C57BL/6 mouse models. The gene expression profiles were detected with Affymetrix microarray and RNA-seq and KEGG (Kyoto Encyclopedia of Genes and Genomes) and CLUE GO analysis was done. The clinical data and transcriptional profiles of melanoma patients from the public database TCGA (The Cancer Genome Atlas) and GEO (Gene Expression Omnibus, GSE32611) were analyzed by Qlucore Omics Explorer. Results NOS1 deletion suppressed the proliferation of melanoma A375 cells in culture, blocked cell cycling at the G0/G1 phase, and decreased the tumor growth in lung metastasis nodes in a B16 melanoma xenograft mouse model. Moreover, NOS1 knockout increased the infiltration of CD3+ immune cells in tumors. The transcriptomics analysis identified 2203 differential expression genes (DEGs) after NOS1 deletion. These DEGs indicated that NOS1 deletion downregulated mostly metabolic functions but upregulated immune response pathways. After inhibiting with NOS1 inhibitor N-PLA, melanoma cells significantly increased the response to IFN\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\upalpha $$\end{document}α by upregulation expression of IFN\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\upalpha $$\end{document}α simulation genes (ISGs), especially the components in innate immune signaling, JAK-STAT, and TOLL-LIKE pathway. Furthermore, these NOS1-regulating immune genes (NOS1-ISGs) worked as a signature to predict poor overall survival and lower response to chemotherapy in melanoma patients. Conclusion These findings provided a transcriptional evidence of NOS1 promotion on tumor growth, which is correlated with metabolic regulation and immune escape in melanoma cells. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03403-w.
Collapse
Affiliation(s)
- Xi Chen
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Zhiwei Zou
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Qianli Wang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Wenwen Gao
- First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450001, China
| | - Sisi Zeng
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Shuangyan Ye
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Pengfei Xu
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Mengqiu Huang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Keyi Li
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Jianping Chen
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Zhuo Zhong
- Guangzhou Hospital of integrated Traditional and West Medicine, Guangzhou, 510800, China
| | - Qianbing Zhang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Bingtao Hao
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China.
| | - Qiuzhen Liu
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China. .,Pingshan District People's Hospital of Shenzhen, Shenzhen, 518118, China.
| |
Collapse
|
3
|
Oliveira-Paula GH, Lacchini R, Tanus-Santos JE. Clinical and pharmacogenetic impact of endothelial nitric oxide synthase polymorphisms on cardiovascular diseases. Nitric Oxide 2017; 63:39-51. [DOI: 10.1016/j.niox.2016.08.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 08/10/2016] [Accepted: 08/24/2016] [Indexed: 12/30/2022]
|
4
|
Kalinowski L, Janaszak-Jasiecka A, Siekierzycka A, Bartoszewska S, Woźniak M, Lejnowski D, Collawn JF, Bartoszewski R. Posttranscriptional and transcriptional regulation of endothelial nitric-oxide synthase during hypoxia: the role of microRNAs. Cell Mol Biol Lett 2016; 21:16. [PMID: 28536619 PMCID: PMC5415778 DOI: 10.1186/s11658-016-0017-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023] Open
Abstract
Understanding the cellular pathways that regulate endothelial nitric oxide (eNOS, NOS3) expression and consequently nitric oxide (NO) bioavailability during hypoxia is a necessary aspect in the development of novel treatments for cardiovascular disorders. eNOS expression and eNOS-dependent NO cellular signaling during hypoxia promote an equilibrium of transcriptional and posttranscriptional molecular mechanisms that belong to both proapoptotic and survival pathways. Furthermore, NO bioavailability results not only from eNOS levels, but also relies on the presence of eNOS substrate and cofactors, the phosphorylation status of eNOS, and the presence of reactive oxygen species (ROS) that can inactivate eNOS. Since both NOS3 levels and these signaling pathways can also be a subject of posttranscriptional modulation by microRNAs (miRNAs), this class of short noncoding RNAs contribute another level of regulation for NO bioavailability. As miRNA antagomirs or specific target protectors could be used in therapeutic approaches to regulate NO levels, either by changing NOS3 mRNA stability or through factors governing eNOS activity, it is critical to understand their role in governing eNOS activity during hypoxa. In contrast to a large number of miRNAs reported to the change eNOS expression during hypoxia, only a few miRNAs modulate eNOS activity. Furthermore, impaired miRNA biogenesis leads to NOS3 mRNA stabilization under hypoxia. Here we discuss the recent studies that define miRNAs’ role in maintaining endothelial NO bioavailability emphasizing those miRNAs that directly modulate NOS3 expression or eNOS activity.
Collapse
Affiliation(s)
- Leszek Kalinowski
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Anna Janaszak-Jasiecka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416 Gdansk, Poland
| | - Anna Siekierzycka
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Sylwia Bartoszewska
- Department of Inorganic Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Woźniak
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Dawid Lejnowski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416 Gdansk, Poland
| | - James F Collawn
- Department of Cell Biology, Developmental, and Integrative, University of Alabama at Birmingham, Birmingham, USA
| | - Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416 Gdansk, Poland
| |
Collapse
|
5
|
Oliveira-Paula GH, Lacchini R, Tanus-Santos JE. Endothelial nitric oxide synthase: From biochemistry and gene structure to clinical implications of NOS3 polymorphisms. Gene 2015; 575:584-99. [PMID: 26428312 DOI: 10.1016/j.gene.2015.09.061] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/10/2015] [Accepted: 09/22/2015] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) is an important vasodilator with a well-established role in cardiovascular homeostasis. While mediator is synthesized from L-arginine by neuronal, endothelial, and inducible nitric oxide synthases (NOS1,NOS3 and NOS2 respectively), NOS3 is the most important isoform for NO formation in the cardiovascular system. NOS3 is a dimeric enzyme whose expression and activity are regulated at transcriptional, posttranscriptional,and posttranslational levels. The NOS3 gene, which encodes NOS3, exhibits a number of polymorphic sites including single nucleotide polymorphisms (SNPs), variable number of tandem repeats (VNTRs), microsatellites, and insertions/deletions. Some NOS3 polymorphisms show functional effects on NOS3 expression or activity, thereby affecting NO formation. Interestingly, many studies have evaluated the effects of functional NOS3 polymorphisms on disease susceptibility and drug responses. Moreover, some studies have investigated how NOS3 haplotypes may impact endogenous NO formation and disease susceptibility. In this article,we carried out a comprehensive review to provide a basic understanding of biochemical mechanisms involved in NOS3 regulation and how genetic variations in NOS3 may translate into relevant clinical and pharmacogenetic implications.
Collapse
Affiliation(s)
- Gustavo H Oliveira-Paula
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
6
|
Yi B, Ozerova M, Zhang GX, Yan G, Huang S, Sun J. Post-Transcriptional Regulation of Endothelial Nitric Oxide Synthase Expression by Polypyrimidine Tract-Binding Protein 1. Arterioscler Thromb Vasc Biol 2015; 35:2153-60. [PMID: 26293469 DOI: 10.1161/atvbaha.115.305750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/06/2015] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Endothelial nitric oxide synthase (eNOS) is an important regulator of vascular function and its expression is regulated at post-transcriptional levels through a yet unknown mechanism. The purpose of this study is to elucidate the post-transcriptional factors regulating eNOS expression and function in endothelium. APPROACHES AND RESULTS To elucidate the molecular basis of tumor necrosis factor (TNF)-α-mediated eNOS mRNA instability, biotinylated eNOS 3'-untranslational region (UTR) was used to purify its associated proteins by RNA affinity chromatography from cytosolic fractions of TNF-α-stimulated human umbilical vein endothelial cells (HUVECs). We identified 2 cytosolic proteins, with molecular weight of 52 and 57 kDa, which specifically bind to eNOS 3'-UTR in response to TNF-α stimulation. Matrix-assisted laser desorption ionization time-of-flight mass spectrometric analysis identified the 57-kDa protein as polypyrimidine tract-binding protein 1 (PTB1). RNA gel mobility shift and UV cross-linking assays demonstrated that PTB1 binds to a UCUU-rich sequence in eNOS 3'-UTR, and the C-terminal half of PTB1 is critical to this interaction. Importantly, PTB1 overexpression leads to decreased activity of luciferase gene fused with eNOS 3'-UTR as well as reduced eNOS expression and activity in human ECs. In HUVECs, we show that TNF-α markedly increased PTB1 expression, whereas adenovirus-mediated PTB1 overexpression decreased eNOS mRNA stability and reduced protein expression and endothelium-dependent relaxation. Furthermore, knockdown of PTB1 substantially attenuated TNF-α-induced destabilization of eNOS transcript and downregulation of eNOS expression. CONCLUSIONS These results indicate that PTB1 is essential for regulating eNOS expression at post-transcriptional levels and suggest a novel therapeutic target for treatment of vascular diseases associated with inflammatory endothelial dysfunction.
Collapse
Affiliation(s)
- Bing Yi
- From the Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (B.Y., M.O., J.S.); Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China (G.Y.); and Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China (G.Z., S.H., J.S.)
| | - Maria Ozerova
- From the Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (B.Y., M.O., J.S.); Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China (G.Y.); and Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China (G.Z., S.H., J.S.)
| | - Guan-Xin Zhang
- From the Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (B.Y., M.O., J.S.); Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China (G.Y.); and Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China (G.Z., S.H., J.S.)
| | - Guijun Yan
- From the Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (B.Y., M.O., J.S.); Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China (G.Y.); and Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China (G.Z., S.H., J.S.)
| | - Shengdong Huang
- From the Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (B.Y., M.O., J.S.); Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China (G.Y.); and Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China (G.Z., S.H., J.S.)
| | - Jianxin Sun
- From the Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (B.Y., M.O., J.S.); Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China (G.Y.); and Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China (G.Z., S.H., J.S.).
| |
Collapse
|
7
|
Galán M, Kassan M, Kadowitz PJ, Trebak M, Belmadani S, Matrougui K. Mechanism of endoplasmic reticulum stress-induced vascular endothelial dysfunction. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:1063-75. [PMID: 24576409 DOI: 10.1016/j.bbamcr.2014.02.009] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/07/2014] [Accepted: 02/17/2014] [Indexed: 12/19/2022]
Abstract
BACKGROUND We recently reported that ER stress plays a key role in vascular endothelial dysfunction during hypertension. In this study we aimed to elucidate the mechanisms by which ER stress induction and oxidative stress impair vascular endothelial function. METHODOLOGY/PRINCIPAL FINDINGS We conducted in vitro studies with primary endothelial cells from coronary arteries stimulated with tunicamycin, 1μg/mL, in the presence or absence of two ER stress inhibitors: tauroursodeoxycholic acid (Tudca), 500μg/mL, and 4-phenylbutyric acid (PBA), 5mM. ER stress induction was assessed by enhanced phosphorylation of PERK and eIF2α, and increased expression of CHOP, ATF6 and Grp78/Bip. The ER stress induction increased p38 MAPK phosphorylation, Nox2/4 mRNA levels and NADPH oxidase activity, and decreased eNOS promoter activity, eNOS expression and phosphorylation, and nitrite levels. Interestingly, the inhibition of p38 MAPK pathway reduced CHOP and Bip expressions enhanced by tunicamycin and restored eNOS promoter activation as well as phosphorylation. To study the effects of ER stress induction in vivo, we used C57BL/6J mice and p47phox(-/-) mice injected with tunicamycin or saline. The ER stress induction in mice significantly impaired vascular endothelium-dependent and independent relaxation in C57BL/6J mice compared with p47phox(-/-) mice indicating NADPH oxidase activity as an intermediate for ER stress in vascular endothelial dysfunction. CONCLUSION/SIGNIFICANCE We conclude that chemically induced ER stress leads to a downstream enhancement of p38 MAPK and oxidative stress causing vascular endothelial dysfunction. Our results indicate that inhibition of ER stress could be a novel therapeutic strategy to attenuate vascular dysfunction during cardiovascular diseases.
Collapse
Affiliation(s)
- Maria Galán
- Department of Physiology, Hypertension and Renal Center of Excellence, Tulane University, 1430 Tulane Ave, New Orleans, LA 70112, USA
| | - Modar Kassan
- Department of Physiology, Hypertension and Renal Center of Excellence, Tulane University, 1430 Tulane Ave, New Orleans, LA 70112, USA
| | - Philip J Kadowitz
- Department of Physiology, Hypertension and Renal Center of Excellence, Tulane University, 1430 Tulane Ave, New Orleans, LA 70112, USA
| | - Mohamed Trebak
- Nanobioscience Constellation, College of Nanoscale Science and Engineering, State University of New York (SUNY), 257 Fuller Rd., Albany, NY 12203, USA
| | - Souad Belmadani
- Department of Physiological Sciences, Eastern Virginia School of Medicine, Norfolk, VA 23501, USA
| | - Khalid Matrougui
- Department of Physiology, Hypertension and Renal Center of Excellence, Tulane University, 1430 Tulane Ave, New Orleans, LA 70112, USA; Department of Physiological Sciences, Eastern Virginia School of Medicine, Norfolk, VA 23501, USA.
| |
Collapse
|
8
|
Enhanced assay of endothelial exocytosis using extracellular matrix components. Anal Biochem 2014; 452:19-24. [PMID: 24561025 DOI: 10.1016/j.ab.2014.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/07/2014] [Accepted: 02/11/2014] [Indexed: 01/05/2023]
Abstract
Vascular inflammation plays a key role in the pathogenesis of atherosclerosis. The first step in vascular inflammation is endothelial exocytosis, in which endothelial granules fuse with the plasma membrane, releasing prothrombotic and proinflammatory messenger molecules. The development of cell culture models to study endothelial exocytosis has been challenging because the factors that modulate exocytosis in vitro are not well understood. Here we report a method for studying endothelial exocytosis that optimizes extracellular matrix components, cell density, and duration of culture. Human umbilical vein endothelial cells plated on collagen I-coated plates and cultured in the confluent state for 7-12 days in low-serum medium showed robust secretion of von Willebrand factor when stimulated with various agonists. This exocytosis assay is rapid and applicable to high-throughput screening.
Collapse
|
9
|
Angiogenic response of endothelial cells to fibronectin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 749:131-51. [PMID: 22695843 DOI: 10.1007/978-1-4614-3381-1_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
10
|
Abstract
Nitric oxide (NO) is a structurally simple, highly versatile molecule that was originally discovered over 30 years ago as an endothelium-derived relaxing factor. In addition to its vasorelaxing effects, NO is now recognized as a key determinant of vascular health, exerting antiplatelet, antithrombotic, and anti-inflammatory properties within the vasculature. This short-lived molecule exerts its inhibitory effect on vascular smooth muscle cells and platelets largely through cyclic guanosine monophosphate-dependent mechanisms, resulting in a multitude of molecular effects by which platelet activation and aggregation are prevented. The biosynthesis of NO occurs via the catalytic activity of NO synthase, an oxidoreductase found in many cell types. NO insufficiency can be attributed to limited substrate/cofactor availability as well as interactions with reactive oxygen species. Impaired NO bioavailability represents the central feature of endothelial dysfunction, a common abnormality found in many vascular diseases. In this review, we present an overview of NO synthesis and biochemistry, discuss the mechanisms of action of NO in regulating platelet and endothelial function, and review the effects of vascular disease states on NO bioavailability.
Collapse
Affiliation(s)
- Richard C Jin
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | |
Collapse
|
11
|
Lazzerini G, Turco SD, Basta G, O’Loghlen A, Zampolli A, Caterina RD. Prominent role of NF-κB in the induction of endothelial activation by endogenous nitric oxide inhibition. Nitric Oxide 2009; 21:184-91. [DOI: 10.1016/j.niox.2009.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2009] [Revised: 07/26/2009] [Accepted: 07/28/2009] [Indexed: 10/20/2022]
|
12
|
Kumar S, Sud N, Fonseca FV, Hou Y, Black SM. Shear stress stimulates nitric oxide signaling in pulmonary arterial endothelial cells via a reduction in catalase activity: role of protein kinase C delta. Am J Physiol Lung Cell Mol Physiol 2009; 298:L105-16. [PMID: 19897742 DOI: 10.1152/ajplung.00290.2009] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous studies have indicated that acute increases in shear stress can stimulate endothelial nitric oxide synthase (eNOS) activity through increased PI3 kinase/Akt signaling and phosphorylation of Ser1177. However, the mechanism by which shear stress activates this pathway has not been adequately resolved nor has the potential role of reactive oxygen species (ROS) been evaluated. Thus, the purpose of this study was to determine if shear-mediated increases in ROS play a role in stimulating Ser1177 phosphorylation and NO signaling in pulmonary arterial endothelial cells (PAEC) exposed to acute increases in shear stress. Our initial studies demonstrated that although shear stress did not increase superoxide levels in PAEC, there was an increase in H2O2 levels. The increases in H2O2 were associated with a decrease in catalase activity but not protein levels. In addition, we found that acute shear stress caused an increase in eNOS phosphorylation at Ser1177 phosphorylation and a decrease in phosphorylation at Thr495. We also found that the overexpression of catalase significantly attenuated the shear-mediated increases in H2O2, phospho-Ser1177 eNOS, and NO generation. Further investigation identified a decrease in PKCdelta activity in response to shear stress, and the overexpression of PKCdelta attenuated the shear-mediated decrease in Thr495 phosphorylation and the increase in NO generation, and this led to increased eNOS uncoupling. PKCdelta overexpression also attenuated Ser1177 phosphorylation through a posttranslational increase in catalase activity, mediated via a serine phosphorylation event, reducing shear-mediated increases in H2O2. Together, our data indicate that shear stress decreases PKCdelta activity, altering the phosphorylation pattern catalase, leading to decreased catalase activity and increased H2O2 signaling, and this in turn leads to increases in phosphorylation of eNOS at Ser1177 and NO generation.
Collapse
Affiliation(s)
- Sanjiv Kumar
- Vascular Biology Center: CB-3210B, Medical College of Georgia, 1459 Laney Walker Blvd., Augusta, GA 30912, USA
| | | | | | | | | |
Collapse
|
13
|
Balligand JL, Feron O, Dessy C. eNOS activation by physical forces: from short-term regulation of contraction to chronic remodeling of cardiovascular tissues. Physiol Rev 2009; 89:481-534. [PMID: 19342613 DOI: 10.1152/physrev.00042.2007] [Citation(s) in RCA: 324] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide production in response to flow-dependent shear forces applied on the surface of endothelial cells is a fundamental mechanism of regulation of vascular tone, peripheral resistance, and tissue perfusion. This implicates the concerted action of multiple upstream "mechanosensing" molecules reversibly assembled in signalosomes recruiting endothelial nitric oxide synthase (eNOS) in specific subcellular locales, e.g., plasmalemmal caveolae. Subsequent short- and long-term increases in activity and expression of eNOS translate this mechanical stimulus into enhanced NO production and bioactivity through a complex transcriptional and posttranslational regulation of the enzyme, including by shear-stress responsive transcription factors, oxidant stress-dependent regulation of transcript stability, eNOS regulatory phosphorylations, and protein-protein interactions. Notably, eNOS expressed in cardiac myocytes is amenable to a similar regulation in response to stretching of cardiac muscle cells and in part mediates the length-dependent increase in cardiac contraction force. In addition to short-term regulation of contractile tone, eNOS mediates key aspects of cardiac and vascular remodeling, e.g., by orchestrating the mobilization, recruitment, migration, and differentiation of cardiac and vascular progenitor cells, in part by regulating the stabilization and transcriptional activity of hypoxia inducible factor in normoxia and hypoxia. The continuum of the influence of eNOS in cardiovascular biology explains its growing implication in mechanosensitive aspects of integrated physiology, such as the control of blood pressure variability or the modulation of cardiac remodeling in situations of hemodynamic overload.
Collapse
Affiliation(s)
- J-L Balligand
- Unit of Pharmacology and Therapeutics, Université catholique de Louvain, Brussels, Belgium.
| | | | | |
Collapse
|
14
|
Youn JY, Wang T, Cai H. An ezrin/calpain/PI3K/AMPK/eNOSs1179 signaling cascade mediating VEGF-dependent endothelial nitric oxide production. Circ Res 2008; 104:50-9. [PMID: 19038867 DOI: 10.1161/circresaha.108.178467] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Calpain was recently reported to mediate vascular endothelial growth factor (VEGF)-induced angiogenesis. In the present study, we investigated detailed molecular mechanisms. VEGF (100 ng/mL) induced a marked increase in endothelial cell production of NO(*), specifically detected by electron spin resonance. This response was abolished by inhibition of calpain with N-acetyl-leucyl-leucyl-norleucinal (ALLN) or Calpeptin. Both also diminished membrane-specific calpain activation by VEGF, which was intriguingly attenuated by silencing ezrin with RNA interference. A rapid membrane colocalization of calpain and ezrin occurred as short as 10 minutes after VEGF stimulation. AKT, AMP-dependent kinase (AMPK), and endothelial nitric oxide synthase (eNOS)(s1179) phosphorylations in VEGF-stimulated endothelial cells were markedly enhanced, which were however significantly attenuated by either ALLN, Calpeptin, or ezrin small interfering RNA, as well as by Wortmannin or compound C (respectively for phosphatidylinositol 3-kinase [PI3K] or AMPK). The latter 3 also abolished VEGF induction of NO(*). These data indicate that AMPK and AKT are both downstream of PI3K and that AKT activation is partially dependent on AMPK. The interrelationship between AMPK and AKT, although known to be individually important in mediating VEGF activation of eNOS, is clearly characterized. Furthermore, AMPK/AKT/eNOS(s1179) was found downstream of a calpain/ezrin membrane interaction. These data no doubt provide new insights into the long mystified signaling gap between VEGF receptors and PI3K/AKT or AMPK-dependent eNOS activation. In view of the well-established significance of VEGF-dependent angiogenesis, these findings might have broad and important implications in cardiovascular pathophysiology.
Collapse
Affiliation(s)
- Ji-Youn Youn
- Division of Molecular Medicine, Cardiovascular Research Laboratories, Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
15
|
Wang X, Chai H, Wang Z, Lin PH, Yao Q, Chen C. Serum amyloid A induces endothelial dysfunction in porcine coronary arteries and human coronary artery endothelial cells. Am J Physiol Heart Circ Physiol 2008; 295:H2399-408. [PMID: 18931033 DOI: 10.1152/ajpheart.00238.2008] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The objective of this study was to determine the effects and mechanisms of serum amyloid A (SAA) on coronary endothelial function. Porcine coronary arteries and human coronary arterial endothelial cells (HCAECs) were treated with SAA (0, 1, 10, or 25 microg/ml). Vasomotor reactivity was studied using a myograph tension system. SAA significantly reduced endothelium-dependent vasorelaxation of porcine coronary arteries in response to bradykinin in a concentration-dependent manner. SAA significantly decreased endothelial nitric oxide (NO) synthase (eNOS) mRNA and protein levels as well as NO bioavailability, whereas it increased ROS in both artery rings and HCAECs. In addition, the activities of internal antioxidant enzymes catalase and SOD were decreased in SAA-treated HCAECs. Bio-plex immunoassay analysis showed the activation of JNK, ERK2, and IkappaB-alpha after SAA treatment. Consequently, the antioxidants seleno-l-methionine and Mn(III) tetrakis-(4-benzoic acid)porphyrin and specific inhibitors for JNK and ERK1/2 effectively blocked the SAA-induced eNOS mRNA decrease and SAA-induced decrease in endothelium-dependent vasorelaxation in porcine coronary arteries. Thus, SAA at clinically relevant concentrations causes endothelial dysfunction in both porcine coronary arteries and HCAECs through molecular mechanisms involving eNOS downregulation, oxidative stress, and activation of JNK and ERK1/2 as well as NF-kappaB. These findings suggest that SAA may contribute to the progress of coronary artery disease.
Collapse
Affiliation(s)
- Xinwen Wang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Mail stop: BCM390, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
16
|
Yan G, You B, Chen SP, Liao JK, Sun J. Tumor necrosis factor-alpha downregulates endothelial nitric oxide synthase mRNA stability via translation elongation factor 1-alpha 1. Circ Res 2008; 103:591-7. [PMID: 18688046 DOI: 10.1161/circresaha.108.173963] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Endothelium-derived nitric oxide (NO) is an important regulator of vascular function. NO is produced by endothelial NO synthase (eNOS), whose expression is downregulated by tumor necrosis factor (TNF)-alpha at the posttranscriptional level. To elucidate the molecular basis of TNF-alpha-mediated eNOS mRNA instability, eNOS 3' untranslated region (3'-UTR) binding proteins were purified by RNA affinity chromatography from cytosolic fractions of TNF-alpha-stimulated human umbilical vein endothelial cells (HUVECs). The formation of 3'-UTR ribonucleoprotein complexes, with molecular weight of 52 and 57 kDa, was increased by TNF-alpha. Matrix-assisted laser desorption ionization time-of-flight mass spectrometric analysis of the 52-kDa protein identified 3 peptides that comprise the peptide sequence of translation elongation factor 1-alpha 1 (eEF1A1). In HUVECs, TNF-alpha rapidly increased eEF1A1 expression, which is maximal after 1 hour and persists for up to 48 hours. RNA gel mobility-shift and UV cross-linking assays indicated that recombinant glutathione S-transferase-eEF1A1 fusion protein specifically binds to a UC-rich sequence in the 3'-UTR of eNOS mRNA. In addition, the domain III of eEF1A1 mediates the binding of eNOS 3'-UTR in eEF1A1. Overexpression of eEF1A1 markedly attenuated the expression of eNOS and luciferase gene fused with eNOS 3'-UTR in both COS-7 cells and bovine aortic endothelial cells (BAECs). Furthermore, adenovirus-mediated overexpression of eEF1A1 increased eNOS mRNA instability, whereas knockdown of eEF1A1 substantially attenuated TNF-alpha-induced destabilization of eNOS mRNA and downregulation of eNOS expression in HUVECs. These results indicate that eEF1A1 is a novel eNOS 3'-UTR binding protein that plays a critical role in mediating TNF-alpha-induced decrease in eNOS mRNA stability.
Collapse
Affiliation(s)
- Guijun Yan
- Department of Cell Biology & Molecular Medicine, UMDNJ-New Jersey Medical School, Newark, NJ 07103, USA
| | | | | | | | | |
Collapse
|
17
|
Soluble CD40 ligand induces endothelial dysfunction in human and porcine coronary artery endothelial cells. Blood 2008; 112:3205-16. [PMID: 18658029 DOI: 10.1182/blood-2008-03-143479] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The purpose of this study was to determine the effects and mechanisms of sCD40L on endothelial dysfunction in both human coronary artery endothelial cells (HCAECs) and porcine coronary artery rings. HCAECs treated with sCD40L showed significant reductions of endothelial nitric oxide synthase (eNOS) mRNA and protein levels, eNOS mRNA stability, eNOS enzyme activity, and cellular NO levels, whereas superoxide anion (O(2)(-)) production was significantly increased. sCD40L enhanced eNOS mRNA 3'UTR binding to cytoplasmic molecules and induced a unique expression pattern of 95 microRNAs. sCD40L significantly decreased mitochondrial membrane potential, and catalase and SOD activities, whereas it increased NADPH oxidase (NOX) activity. sCD40L increased phosphorylation of MAPKs p38 and ERK1/2 as well as IkappaBalpha and enhanced NF-kappaB nuclear translocation. In porcine coronary arteries, sCD40L significantly decreased endothelium-dependent vasorelaxation and eNOS mRNA levels, whereas it increased O(2)(-) levels. Antioxidant seleno-l-methionine; chemical inhibitors of p38, ERK1/2, and mitochondrial complex II; as well as dominant negative mutant forms of IkappaBalpha and NOX4 effectively blocked sCD40L-induced eNOS down-regulation in HCAECs. Thus, sCD40L reduces eNOS levels, whereas it increases oxidative stress through the unique molecular mechanisms involving eNOS mRNA stability, 3'UTR-binding molecules, microRNAs, mitochondrial function, ROS-related enzymes, p38, ERK1/2, and NF-kappaB signal pathways in endothelial cells.
Collapse
|
18
|
Chatterjee A, Black SM, Catravas JD. Endothelial nitric oxide (NO) and its pathophysiologic regulation. Vascul Pharmacol 2008; 49:134-40. [PMID: 18692595 DOI: 10.1016/j.vph.2008.06.008] [Citation(s) in RCA: 164] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Accepted: 06/16/2008] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO) is a gaseous lipophilic free radical generated by three distinct isoforms of nitric oxide synthases (NOS), type 1 or neuronal (nNOS), type 2 or inducible (iNOS) and type 3 or endothelial NOS (eNOS). Expression of eNOS is altered in many types of cardiovascular disease, such as atherosclerosis, diabetes and hypertension. The ubiquitous chaperone heat shock protein 90 (hsp90) associates with NOS and is important for its proper folding and function. Current studies point toward a therapeutic potential by modulating hsp90-NOS association in various vascular diseases. Here we review the transcriptional regulation of endothelial NOS and factors affecting eNOS activity and function, as well as the important vascular pathologies associated with altered NOS function, focusing on the regulatory role of hsp90 and other factors in NO-associated pathogenesis of these diseases.
Collapse
Affiliation(s)
- Anuran Chatterjee
- Pulmonary Vascular Disease Program, Vascular Biology Center, Medical College of Georgia, Augusta, Georgia 30912-2500, USA
| | | | | |
Collapse
|
19
|
Ihara H, Tsutsuki H, Ida T, Kozaki S, Tsuyama S, Moss J. Alternative polyadenylation sites of human endothelial nitric oxide synthase mRNA. Biochem Biophys Res Commun 2007; 363:146-52. [PMID: 17825792 PMCID: PMC3021985 DOI: 10.1016/j.bbrc.2007.08.118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Accepted: 08/22/2007] [Indexed: 11/20/2022]
Abstract
The mRNA 3'-untranslated region (3'-UTR) has been shown to have important roles in the regulation of mRNA function. In this study, we investigated the human endothelial nitric oxide synthase (eNOS) 3'-UTR to evaluate its potential regulatory role. 3'-RACE analysis revealed that the human eNOS mRNA has multiple alternative polyadenylation sites. Apart from the proximal site (418bp downstream of the stop codon), we identified two additional distal sites approximately 770 and 1478bp downstream of the stop codon. In addition, Northern analysis showed that the usage of these sites differed among human tissues. Further, amounts of these eNOS mRNAs were changed during growth of cultured human aortic endothelial cells; mRNAs with long 3'-UTRs decreased more rapidly than total mRNA, as cells approached confluency. Thus, the 3'-UTRs of human eNOS results from alternative polyadenylation sites and differ across tissues and during cell growth.
Collapse
Affiliation(s)
- Hideshi Ihara
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, 1-1 Gakuen-cho, Sakai, Osaka 599-8531, Japan.
| | | | | | | | | | | |
Collapse
|
20
|
Boudko DY. Bioanalytical profile of the L-arginine/nitric oxide pathway and its evaluation by capillary electrophoresis. J Chromatogr B Analyt Technol Biomed Life Sci 2007; 851:186-210. [PMID: 17329176 PMCID: PMC2040328 DOI: 10.1016/j.jchromb.2007.02.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2006] [Revised: 01/30/2007] [Accepted: 02/06/2007] [Indexed: 02/07/2023]
Abstract
This review briefly summarizes recent progress in fundamental understanding and analytical profiling of the L-arginine/nitric oxide (NO) pathway. It focuses on key analytical references of NO actions and the experimental acquisition of these references in vivo, with capillary electrophoresis (CE) and high-performance capillary electrophoresis (HPCE) comprising one of the most flexible and technologically promising analytical platform for comprehensive high-resolution profiling of NO-related metabolites. Another aim of this review is to express demands and bridge efforts of experimental biologists, medical professionals and chemical analysis-oriented scientists who strive to understand evolution and physiological roles of NO and to develop analytical methods for use in biology and medicine.
Collapse
Affiliation(s)
- Dmitri Y Boudko
- The Whitney Laboratory for Marine Bioscience, 9505 Ocean Shore Blvd., St. Augustine, FL 32080, USA.
| |
Collapse
|
21
|
Weinberg JB, Fermor B, Guilak F. Nitric oxide synthase and cyclooxygenase interactions in cartilage and meniscus: relationships to joint physiology, arthritis, and tissue repair. Subcell Biochem 2007; 42:31-62. [PMID: 17612045 DOI: 10.1007/1-4020-5688-5_2] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Rheumatoid arthritis and osteoarthritis are painful and debilitating diseases with complex pathophysiology. There is growing evidence that pro-inflammatory cytokines (e.g., interleukin-1 and tumor necrosis factor alpha) and mediators (e.g., prostaglandins, leukotrienes, and nitric oxide) play critical roles in the development and perpetuation of tissue inflammation and damage in joint tissues such as articular cartilage and meniscus. While earlier studies have generally focused on cells of the synovium (especially macrophages), there is increasing evidence that chondrocytes and meniscal cells actively contribute to inflammatory processes. In particular, it is now apparent that mechanical forces engendered by joint loading are transduced to biological signals at the cellular level and that these signals modulate gene expression and biochemical processes. Here we give an overview of the interplay of cytokines and mechanical stress in the production of cyclooxygenases and prostaglandins; lipoxygenases and leukotrienes; and nitric oxide synthases and nitric oxide in arthritis, with particular focus on the interactions of these pathways in articular cartilage and meniscus.
Collapse
MESH Headings
- Animals
- Arthritis, Rheumatoid/enzymology
- Arthritis, Rheumatoid/physiopathology
- Cartilage, Articular/enzymology
- Cartilage, Articular/pathology
- Cartilage, Articular/physiopathology
- Chondrocytes/enzymology
- Chondrocytes/pathology
- Gene Expression Regulation, Enzymologic
- Humans
- Inflammation/enzymology
- Inflammation/pathology
- Inflammation Mediators/metabolism
- Macrophages/enzymology
- Macrophages/pathology
- Menisci, Tibial/enzymology
- Menisci, Tibial/pathology
- Menisci, Tibial/physiopathology
- Nitric Oxide Synthase/biosynthesis
- Osteoarthritis, Knee/enzymology
- Osteoarthritis, Knee/pathology
- Osteoarthritis, Knee/physiopathology
- Prostaglandin-Endoperoxide Synthases/biosynthesis
- Regeneration
- Signal Transduction
- Stress, Mechanical
- Synovial Membrane/enzymology
- Synovial Membrane/pathology
Collapse
Affiliation(s)
- J Brice Weinberg
- Department of Medicine, Division of Hematology-Oncology, VA and Duke University Medical Centers, Durham, North Carolina 27705, USA.
| | | | | |
Collapse
|
22
|
Dudzinski D, Michel T. Life history of eNOS: partners and pathways. Cardiovasc Res 2007; 75:247-60. [PMID: 17466957 PMCID: PMC2682334 DOI: 10.1016/j.cardiores.2007.03.023] [Citation(s) in RCA: 303] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Revised: 03/26/2007] [Accepted: 03/28/2007] [Indexed: 02/07/2023] Open
Abstract
The complex regulation of eNOS (endothelial nitric oxide synthase) in cardiovascular physiology occurs at multiple stages. eNOS mRNA levels are controlled both at the transcriptional and post-transcriptional phases, and epigenetic mechanisms appear to modulate tissue-specific eNOS expression. The eNOS enzyme reversibly associates with a diverse family of protein partners that regulate eNOS sub-cellular localization, catalytic function, and biological activity. eNOS enzyme activity and sub-cellular localization are intimately controlled by post-translational modifications including phosphorylation, nitrosylation, and acylation. The multiple extra-cellular stimuli affecting eNOS function coordinate their efforts through these key modifications to dynamically control eNOS and NO bioactivity in the vessel wall. This review will focus on the biochemical partners and perturbations of the eNOS protein as this vital enzyme undergoes modulation by diverse signal transduction pathways in the vascular endothelium.
Collapse
Affiliation(s)
- David Dudzinski
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Thomas Michel
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, p: (617) 732-7376, f: (617) 732-5132, e:
| |
Collapse
|
23
|
Fish JE, Matouk CC, Yeboah E, Bevan SC, Khan M, Patil K, Ohh M, Marsden PA. Hypoxia-inducible expression of a natural cis-antisense transcript inhibits endothelial nitric-oxide synthase. J Biol Chem 2007; 282:15652-66. [PMID: 17403686 DOI: 10.1074/jbc.m608318200] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The destabilization of endothelial nitric-oxide synthase (eNOS) mRNA in hypoxic endothelial cells may be important in the etiology of vascular diseases, such as pulmonary hypertension. Recently, an overlapping antisense transcript to eNOS/NOS3 was implicated in the post-transcriptional regulation of eNOS. We demonstrate here that expression of sONE, also known as eNOS antisense (NOS3AS) or autophagy 9-like 2 (APG9L2), is robustly induced by hypoxia or functional deficiency of von Hippel-Lindau protein. sONE is also up-regulated in the aortas of hypoxic rats. In hypoxic endothelial cells, sONE expression negatively correlates with eNOS expression. Blocking the hypoxic induction of sONE by RNA interference attenuates the fall in both eNOS RNA and protein. We provide evidence that the induction of sONE primarily involves transcript stabilization rather than increased transcriptional activity and is von Hippel-Lindaubut not hypoxia-inducible factor 2alpha-dependent. We also demonstrate that sONE transcripts are enriched in the nucleus of normoxic cells and that hypoxia promotes an increase in the level of cytoplasmic and polyribosome-associated, sONE mRNA. The finding that eNOS expression can be regulated by an overlapping cis-antisense transcript in a stimulus-dependent fashion provides evidence that sense/antisense interactions may play a previously unappreciated role in vascular disease pathogenesis.
Collapse
Affiliation(s)
- Jason E Fish
- Department of Medical Biophysics, St. Michael's Hospital, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Eberhardt W, Doller A, Akool ES, Pfeilschifter J. Modulation of mRNA stability as a novel therapeutic approach. Pharmacol Ther 2007; 114:56-73. [PMID: 17320967 DOI: 10.1016/j.pharmthera.2007.01.002] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 01/10/2007] [Accepted: 01/10/2007] [Indexed: 12/21/2022]
Abstract
During the last decade evidence has accumulated that modulation of mRNA stability plays a central role in cellular homeostasis, including cell differentiation, proliferation and adaptation to external stimuli. The functional relevance of posttranscriptional gene regulation is highlighted by many pathologies, wherein occurrence tightly correlates with a dysregulation in mRNA stability, including chronic inflammation, cardiovascular diseases and cancer. Most commonly, the cis-regulatory elements of mRNA decay are represented by the adenylate- and uridylate (AU)-rich elements (ARE) which are specifically bound by trans-acting RNA binding proteins, which finally determine whether mRNA decay is delayed or facilitated. Regulation of mRNA decay by RNA stabilizing and RNA destabilizing factors is furthermore controlled by different intrinsic and environmental stimuli. The modulation of mRNA binding proteins, therefore, illuminates a promising approach for the pharmacotherapy of those key pathologies mentioned above and characterized by a posttranscriptional dysregulation. Most promisingly, intracellular trafficking of many of the mRNA stability regulating factors is, in turn, regulated by some major signaling pathways, including the mitogen-activated protein kinase (MAPK) cascade, the AMP-activated kinase (AMPK) and the protein kinase (PK) C (PKC) family. In this review, we present timely examples of genes regulated by mRNA stability with a special focus on signaling pathways involved in the ARE-dependent mRNA decay. A better understanding of these processes may form the basis for the development of novel therapeutics to treat major human diseases.
Collapse
Affiliation(s)
- Wolfgang Eberhardt
- Pharmazentrum frankfurt/ZAFES, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany.
| | | | | | | |
Collapse
|
25
|
Misquitta CM, Chen T, Grover AK. Control of protein expression through mRNA stability in calcium signalling. Cell Calcium 2006; 40:329-46. [PMID: 16765440 DOI: 10.1016/j.ceca.2006.04.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2006] [Revised: 03/22/2006] [Accepted: 04/12/2006] [Indexed: 01/14/2023]
Abstract
Specific sequences (cis-acting elements) in the 3'-untranslated region (UTR) of RNA, together with stabilizing and destabilizing proteins (trans-acting factors), determine the mRNA stability, and consequently, the level of expression of several proteins. Such interactions were discovered initially for short-lived mRNAs encoding cytokines and early genes like c-jun and c-myc. However, they may also determine the fate of more stable mRNAs in a tissue and disease-dependent manner. The interactions between the cis-acting elements and the trans-acting factors may also be modulated by Ca(2+) either directly or via a control of the phosphorylation status of the trans-acting factors. We focus initially on the basic concepts in mRNA stability with the trans-acting factors AUF1 (destabilizing) and HuR (stabilizing). Sarco/endoplasmic reticulum Ca(2+) pumps, SERCA2a (cardiac and slow twitch muscles) and SERCA2b (most cells including smooth muscle cells), are pivotal in Ca(2+) mobilization during signal transduction. SERCA2a and SERCA2b proteins are encoded by relatively stable mRNAs that contain cis-acting stability determinants in their 3'-regions. We present several pathways where 3'-UTR mediated mRNA decay is key to Ca(2+) signalling: SERCA2a and beta-adrenergic receptors in heart failure, renin-angiotensin system, and parathyroid hormones. Other examples discussed include cytokines vascular endothelial growth factor, endothelin and endothelial nitric oxide synthase. Roles of Ca(2+) and Ca(2+)-binding proteins in mRNA stability are also discussed. We anticipate that these novel modes of control of protein expression will form an emerging area of research that may explore the central role of Ca(2+) in cell function during development and in disease.
Collapse
Affiliation(s)
- Christine M Misquitta
- Banting and Best Department of Medical Research, 10th floor Donnelly CCBR, University of Toronto, 160 College Street, Toronto, Ont., Canada M5S 3E1
| | | | | |
Collapse
|
26
|
Searles CD. Transcriptional and posttranscriptional regulation of endothelial nitric oxide synthase expression. Am J Physiol Cell Physiol 2006; 291:C803-16. [PMID: 16738003 DOI: 10.1152/ajpcell.00457.2005] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The ability of the endothelium to produce nitric oxide is essential to maintenance of vascular homeostasis; disturbance of this ability is a major contributor to the pathogenesis of vascular disease. In vivo studies have demonstrated that expression of endothelial nitric oxide synthase (eNOS) is vital to endothelial function and have led to the understanding that eNOS expression is subject to modest but significant degrees of regulation. Subsequently, numerous physiological and pathophysiological stimuli have been identified that modulate eNOS expression via mechanisms that alter steady-state eNOS mRNA levels. These mechanisms involve changes in the rate of eNOS gene transcription (transcriptional regulation) and alteration of eNOS mRNA processing and stability (posttranscriptional regulation). In cultured endothelial cells, shear stress, transforming growth factor-beta1, lysophosphatidylcholine, cell growth, oxidized linoleic acid, 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, and hydrogen peroxide have been shown to increase eNOS expression. In contrast, tumor necrosis factor-alpha, hypoxia, lipopolysaccaride, thrombin, and oxidized LDL can decrease eNOS mRNA levels. For many of these stimuli, both transcriptional and posttranscriptional mechanisms contribute to regulation of eNOS expression. Recent studies have begun to further define signaling pathways responsible for changes in eNOS expression and have characterized cis- and trans-acting regulatory elements. In addition, a role has been identified for epigenetic control of eNOS mRNA levels. This review will discuss transcriptional and posttranscriptional regulation of eNOS with emphasis on the molecular mechanisms that have been identified for these processes.
Collapse
Affiliation(s)
- Charles D Searles
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
27
|
Abstract
Unlike other types of cancer, tumors of the breast are greatly influenced by steroid hormones. The effect of estrogen and progesterone depends on the presence of their specific receptors and these constitute important parameters in determining the aggressiveness of the tumor, the feasibility of certain therapies and the prediction of relapse. The molecular mechanisms of steroid hormone action have not been fully elucidated but recent findings implicate the nitric oxide (NO) pathway in some of these effects. Both hormones can regulate the nitric oxide synthases (NOS) and, in turn, the NO produced has profound consequences on tumor cell homeostasis. On one hand, estrogen increases the activity of endothelial NOS (eNOS or NOSIII), while progesterone activates inducible NOS (iNOS or NOSII) expression. The data presented suggest that the low levels of NO produced by NOSIII mediate the proliferative effect of estrogen. On the other hand, the increase in apoptosis in response to progesterone could implicate the high levels of NO produced by induction of NOSII expression. Understanding of the mechanisms and interactions of steroid hormones with the NO pathway could lead to the development of new approaches and strategies for the effective treatment of breast cancer.
Collapse
Affiliation(s)
- Alena Pance
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK.
| |
Collapse
|
28
|
Andreeva AV, Vaiskunaite R, Kutuzov MA, Profirovic J, Skidgel RA, Voyno-Yasenetskaya T. Novel mechanisms of G protein-dependent regulation of endothelial nitric-oxide synthase. Mol Pharmacol 2006; 69:975-82. [PMID: 16326932 DOI: 10.1124/mol.105.018846] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Endothelial nitric-oxide synthase (eNOS) plays a crucial role in the regulation of a variety of cardiovascular and pulmonary functions in both normal and pathological conditions. Multiple signaling inputs, including calcium, caveolin-1, phosphorylation by several kinases, and binding to the 90-kDa heat shock protein (Hsp90), regulate eNOS activity. Here, we report a novel mechanism of G protein-dependent regulation of eNOS. We demonstrate that in mammalian cells, the alpha subunit of heterotrimeric G12 protein (G alpha12) can form a complex with eNOS in an activation- and Hsp90-independent manner. Our data show that G alpha12 does not affect eNOS-specific activity, but it strongly enhances total eNOS activity by increasing cellular levels of eNOS. Experiments using inhibition of protein or mRNA synthesis show that G alpha12 increases the expression of eNOS by increasing half-life of both eNOS protein and eNOS mRNA. Small interfering RNA-mediated depletion of endogenous G alpha12 decreases eNOS levels. A quantitative correlation can be detected between the extent of down-regulation of G alpha12 and eNOS in endothelial cells after prolonged treatment with thrombin. G protein-dependent increase of eNOS expression represents a novel mechanism by which heterotrimeric G proteins can regulate the activity of downstream signaling molecules.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Enzyme Activation
- Enzyme Stability
- GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors
- GTP-Binding Protein alpha Subunits, G12-G13/genetics
- GTP-Binding Protein alpha Subunits, G12-G13/metabolism
- HSP90 Heat-Shock Proteins/metabolism
- Humans
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- RNA Stability
- RNA, Messenger/analysis
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/pharmacology
- Thrombin/pharmacology
Collapse
Affiliation(s)
- Alexandra V Andreeva
- Department of Pharmacology, University of Illinois, College of Medicine, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
29
|
Cale JM, Tsoi SC, Toppe M, Grummer MA, Ochiai M, Magness RR, Bird IM. Molecular cloning of ovine endothelial nitric oxide synthase and expression in COS-7 cells. ACTA ACUST UNITED AC 2005; 12:156-68. [PMID: 15784500 DOI: 10.1016/j.jsgi.2004.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
While studies of human and bovine endothelial nitric oxide synthase (eNOS) demonstrate activation by Ca(2+)/calmodulin, recent progress demonstrates that eNOS phosphorylation can alter sensitivity to intracellular free calcium ([Ca(2+)](i)). The sheep, however, is widely used as a model for cardiovascular adaptation to pregnancy and ovine uterine artery endothelial cell (UAEC) eNOS undergoes pregnancy-specific (P) enhancement of activity associated with increased Ca(2+) and protein kinase signaling in response to a number of agonists, including adenosine triphosphate (ATP). The degree of homology between the ovine and human full-length cDNAs was not previously known and yet is necessary to determine the validity in using an ovine model to study human physiology. The objectives of this study were to isolate and validate the clone of ovine eNOS cDNA and investigate ovine eNOS activation when expressed in COS-7 cells. The ovine eNOS cDNA has high homology to published human and bovine sequences and shares identity with the bovine amino acid sequence. When ovine eNOS was transiently expressed in COS-7 cells (COS-7/oeNOS), A23187 increased specific catalytic activity in a dose- and time-dependent manner. A23187-stimulated activation of eNOS was, however, also accompanied by phosphorylation of eNOS S1179 and dephosphorylation of T497, demonstrating that an increase in [Ca(2+)](i) may not be the sole mechanism of activation. The physiologic relevance of this was further underscored by the finding that ATP dose-dependently increased peak [Ca(2+)](i) and eNOS activity in COS-7/oeNOS, but also increased eNOS p-S1179 and decreased p-T497. This finding was similar to those in ovine P-UAEC treated with the Ca(2+)-mobilizing agonist ATP, wherein activation of eNOS was again concomitant with a rise p-S1179 as well as a slight decrease in p-T497. In conclusion, we describe the full-length ovine eNOS cDNA sequence and show that both physiologic and nonphysiologic calcium-mobilizing agents, which activate ovine eNOS in COS-7 and P-UAEC, do so in association with changes in eNOS phosphorylation. Given this information we can now begin to dissect the relationship between Ca(2+) elevation and specific phosphorylation events in eNOS activation in the ovine model, and thereby gain insight into the possible basis for pregnancy-related dysfunction.
Collapse
Affiliation(s)
- Jacqueline M Cale
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Stangl V, Lorenz M, Meiners S, Ludwig A, Bartsch C, Moobed M, Vietzke A, Kinkel HT, Baumann G, Stangl K. Long‐term up‐regulation of eNOS and improvement of endothelial function by inhibition of the ubiquitin–proteasome pathway. FASEB J 2004; 18:272-9. [PMID: 14769821 DOI: 10.1096/fj.03-0054com] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The ubiquitin-proteasome system is the major pathway for intracellular protein degradation in eukaryotic cells. Endothelial nitric oxide synthase (eNOS) is the key enzyme of vascular homeostasis involved in the pathophysiology of several cardiovascular diseases. The aim of our study was to investigate whether eNOS expression and activity are regulated by the proteasome. Bovine pulmonary artery endothelial cells (CPAE cells) were treated with the proteasome inhibitor MG132. MG132 (50-250 nmol/L) dose-dependently increased mRNA and protein levels of eNOS. Comparable results were obtained with other specific proteasome inhibitors, whereas the nonproteasomal calpain and cathepsin inhibitor ALLM had no effect. Efficacy of proteasome inhibition was evidenced by accumulation of poly-ubiquitinylated proteins and by measuring proteasomal activity in cell extracts. Cycloheximide prevented up-regulation of eNOS protein, indicating that post-translational stabilization of eNOS is not involved. eNOS activity was increased up to 2.8-fold (MG132 100 nmol/L, 48 h). Incubation of rat aortic rings with MG132 significantly enhanced endothelial-dependent vasorelaxation. Single MG132 treatment (100 nmol/L) induced long-term effects in CPAE cells, with increases of eNOS protein and activity for up to 10 days. Our results indicate that low-dose proteasome inhibition enhances eNOS expression and activity, and improves endothelial function.
Collapse
Affiliation(s)
- Verena Stangl
- Medizinische Klinik mit Schwerpunkt Kardiologie, Angiologie, Pneumologie, Humboldt-Universität Berlin, Charité, Campus Mitte, Schumannstr. 20-21, D-10117 Berlin, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Coulet F, Nadaud S, Agrapart M, Soubrier F. Identification of hypoxia-response element in the human endothelial nitric-oxide synthase gene promoter. J Biol Chem 2003; 278:46230-40. [PMID: 12963737 DOI: 10.1074/jbc.m305420200] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The human endothelial nitric-oxide synthase gene (heNOS) is constitutively expressed in endothelial cells, and its expression is induced under hypoxia. The goal of this study was to search for regulatory elements of the endothelial nitric-oxide synthase (eNOS) gene responsive to hypoxia. Levels of eNOS mRNA, measured by real time reverse transcriptase-PCR analysis, were increased, and heNOS promoter activity was enhanced by hypoxia as compared with normoxia control experiments. Promoter truncation followed by footprint analysis allowed the mapping and identification of the hypoxia-responsive elements at position -5375 to -5366, closely related to hypoxia-inducible factor (HIF)-responsive element (HRE). To test whether known HIF-1 and HIF-2 are involved in hypoxia-induced heNOS promoter activation, HMEC-1 and HUVEC were transiently transfected with HIF-1alpha and HIF-1beta or HIF-2alpha and HIF-1beta expression vectors. Exogenous HIF-2 markedly increased luciferase reporter activity driven by the heNOS promoter in its native location. The induction of luciferase was conserved with the antisense construct and was increased in cotransfection experiments when this fragment was cloned 5' to the proximal 785-bp fragment of the eNOS promoter. Deletion analysis and site-directed mutagenesis demonstrated that the two contiguous HIF consensus binding sites spanning bp -5375 to -5366 relative to the transcription start site were both functional for heNOS promoter activity induction by hypoxia and by HIF-2 overexpression. In conclusion, we demonstrate that heNOS is a hypoxia-inducible gene, whose transcription is stimulated through HIF-2 interaction with two contiguous HRE sites located at -5375 to -5366 of the heNOS promoter.
Collapse
Affiliation(s)
- Florence Coulet
- INSERM, Unit 525, Université Pierre et Marie Curie, Faculté de Médecine Pitié-Salpêtrière, 91 Boulevard de l'Hôpital, 75013 Paris, France
| | | | | | | |
Collapse
|
32
|
Bayraktutan U. Effects of angiotensin II on nitric oxide generation in growing and resting rat aortic endothelial cells. J Hypertens 2003; 21:2093-101. [PMID: 14597853 DOI: 10.1097/00004872-200311000-00019] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES To assess the effects of angiotensin II (ang II) and its receptors on nitric oxide (NO) production and endothelial NO synthase (eNOS) activity and expression with respect to rat aortic endothelial cell (RAEC) growth. To also assess whether an intact endothelium is required for ang II activity. METHODS RAEC were treated with different doses of ang II, Ca(2+) ionophore A23187, valsartan (an AT(1) receptor inhibitor) or PD-123319 (an AT(2) receptor inhibitor) alone or in combination for 24 h before measuring nitrite levels by Griess reaction as an index of NO production and eNOS activity by L-[3H]-arginine to L-[3H]-citrulline conversion assay. eNOS mRNA and protein expressions were determined by Northern and Western analyses, respectively. The requirement of endothelium for ang II-mediated relaxant/contractile effects was investigated by isometric tension studies. RESULTS NO production and eNOS activity/expression were almost two-fold greater in proliferating RAEC. Ang II or Ca(2+) ionophore A23187 enhanced NO production in proliferating and confluent RAEC without altering the fold-difference in basal NO release. Both valsartan and PD-123319 significantly diminished NO production in RAEC treated with ang II but not Ca(2+) ionophore A23187 while NG-nitro-L-arginine (L-NNA, 10 micromol/l) equally decreased NO generation in response to both stimulators. L-NNA, valsartan and PD-123319 also abolished endothelium-dependent vasorelaxant responses to ACh and Ca(2+) ionophore A23187 in the presence of ang II. Sodium nitroprusside (SNP), a NO donor, increased endothelium-independent vasorelaxant responses that were augmented by valsartan but not L-NNA or PD-123319 in the presence of ang II. CONCLUSIONS Ang II induces vascular NO production through endothelial AT(1) and AT(2)-receptors. This may be beneficial in counterbalancing its vasoconstrictor effect on vascular smooth muscle cells.
Collapse
Affiliation(s)
- Ulvi Bayraktutan
- Department of Medicine, Institute of Clinical Science, University Belfast, Belfast, UK.
| |
Collapse
|
33
|
Abstract
This article has focused on the influence of NO. on vascular homeostasis. Vascular tone, however, is also influenced by other vasoactive factors released by the endothelium, including the endothelial-derived hyperpolarizing factors, prostacyclin, and vasoconstrictor factors. There is also abundant evidence that these factors are altered by pathophysiologic states, although the mechanisms responsible are not as well understood as they seem to be for the NO. system. There is now evidence that several endothelial-derived hyperpolarizing factors may exist. One is almost certainly the cytochrome p450 metabolite of arachidonic acid, epoxyeicosatrienoic acid (EET) [92], whereas another is likely H2O2, which stimulates potassium channel opening in a fashion similar to the EET [93]. EET has anti-inflammatory properties, whereas H2O2 may potentially enhance inflammation and promote vascular hypertrophy. Thus, two factors released by the endothelium with similar acute effects on the vascular smooth muscle may have very different long-term consequences in terms of protecting against or promoting vascular disease. During the past two decades, physicians have gained a substantial understanding of the L-arginine/eNOS/NO. pathway and how this modulates vascular reactivity. Further, physicians now are aware that this process is altered by many risk factors for atherosclerosis and have begun to understand how these disorders alter NO. production and bioavailability. These abnormalities are likely multifactorial and physicians are beginning to understand how they can be corrected. An exciting aspect of endothelial function is that it has prognostic significance above and beyond the traditional risk factors for atherosclerosis. Several studies now have shown that individuals with intact endothelial function in either the forearm or the coronary circulation have a low incidence of events during follow-up periods, whereas those individuals with abnormal endothelial function have a high incidence of major cardiovascular events [94-96]. Because of the complexity of abnormalities that underlie endothelial dysfunction, there are various therapeutic targets that may have to be addressed to improve endothelial function and ultimately improve prognosis in these individuals.
Collapse
Affiliation(s)
- David G Harrison
- Division of Cardiology, Emory University School of Medicine, 1639 Pierce Drive, Room 319 WMRB, Atlanta, GA 30322, USA.
| | | |
Collapse
|
34
|
Bayraktutan U, Ulker S. Effects of Angiotensin II on Nitric Oxide Generation in Proliferating and Quiescent Rat Coronary Microvascular Endothelial Cells. Hypertens Res 2003; 26:749-57. [PMID: 14620932 DOI: 10.1291/hypres.26.749] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Nitric oxide (NO) and the mitogenic peptide angiotensin II (Ang II) have been implicated in endothelial cell growth. However, the putative relationship between these two opposing agents with respect to endothelial cell growth remains unknown. In this study, proliferating and confluent rat coronary microvascular endothelial cells (CMEC) were treated with different doses of Ang II, Ca2+ ionophore A23187, or valsartan (an Ang II type 1 (AT1) receptor inhibitor) alone or in combination for 24 h before measuring the nitrite levels as an index of NO generation. NO production and endothelial NO synthase (eNOS) mRNA/protein expression were found to be 3-fold greater in proliferating vs. quiescent CMEC. Treatments of CMEC with Ang II or Ca2+ ionophore A23187 equally increased NO production without altering the fold-difference in the basal release of NO from proliferating vs. confluent CMEC. Valsartan abolished NO production in CMEC treated with Ang II but not Ca2+ ionophore A23187. Treatments of endothelium-intact vascular rings with Ang II (1 nmol/l to 10 micromol/l) plus valsartan or PD-123319, an Ang II type 2 (AT2) receptor inhibitor, attenuated vascular responses to acetylcholine in an Ang II dose-dependent manner. In these rings, phenylephrine produced significant increases in contractile responses only at nmol/l concentrations of Ang II. In contrast, pharmacological and mechanical inactivation of endothelium enhanced contractile responses to phenylephrine at micromol/I concentrations of Ang II. These data demonstrate that Ang II stimulates NO production in CMEC in both an AT1- and an AT2 receptor-regulated manner, and that this stimulation of NO may be beneficial in counterbalancing the direct vasoconstrictor effect of Ang II on underlying smooth muscle cells.
Collapse
Affiliation(s)
- Ulvi Bayraktutan
- Department of Medicine, Institute of Clinical Science, Block B, Queen's University Belfast, Grosvenor Road, Belfast BT12 6BJ, UK.
| | | |
Collapse
|
35
|
Fleming I, Busse R. Molecular mechanisms involved in the regulation of the endothelial nitric oxide synthase. Am J Physiol Regul Integr Comp Physiol 2003; 284:R1-12. [PMID: 12482742 DOI: 10.1152/ajpregu.00323.2002] [Citation(s) in RCA: 606] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The endothelial nitric oxide synthase (eNOS), the expression of which is regulated by a range of transcriptional and posttranscriptional mechanisms, generates nitric oxide (NO) in response to a number of stimuli. The physiologically most important determinants for the continuous generation of NO and thus the regulation of local blood flow are fluid shear stress and pulsatile stretch. Although eNOS activity is coupled to changes in endothelial cell Ca(2+) levels, an increase in Ca(2+) alone is not sufficient to affect enzyme activity because the binding of calmodulin (CaM) and the flow of electrons from the reductase to the oxygenase domain of the enzyme is dependent on protein phosphorylation and dephosphorylation. Two amino acids seem to be particularly important in regulating eNOS activity and these are a serine residue in the reductase domain (Ser(1177)) and a threonine residue (Thr(495)) located within the CaM-binding domain. Simultaneous alterations in the phosphorylation of Ser(1177) and Thr(495) in response to a variety of stimuli are regulated by a number of kinases and phosphatases that continuously associate with and dissociate from the eNOS signaling complex. eNOS associated proteins, such as caveolin, heat shock protein 90, eNOS interacting protein, and possibly also motor proteins provide the scaffold for the formation of the protein complex as well as its intracellular localization.
Collapse
Affiliation(s)
- Ingrid Fleming
- Institut für Kardiovaskuläre Physiologie, J. W. Goethe-Universität, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany.
| | | |
Collapse
|
36
|
Napoli C, Lerman LO, de Nigris F, Loscalzo J, Ignarro LJ. Glycoxidized low-density lipoprotein downregulates endothelial nitricoxide synthase in human coronary cells. J Am Coll Cardiol 2002; 40:1515-22. [PMID: 12392844 DOI: 10.1016/s0735-1097(02)02306-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES We examined the hypothesis that low-density lipoprotein (LDL) that is both oxidized and glycosylated potently downregulates the expression of endothelial nitric oxide synthase III (NOSIII) in human coronary endothelial cells. BACKGROUND Diabetes mellitus is accompanied by both oxidation and glycosylation of LDL, but the potential interaction of these processes or the pathophysiologic effects of these modified lipoproteins on arteries are poorly understood. METHODS Low-density lipoprotein was glycoxidized in vitro, and Western and Northern blot analyses were used to investigate NOSIII expression in human coronary endothelial cells. Nitric oxide (NO) bioactivity was represented by both basal and bradykinin-stimulated cellular cyclic guanosine monophosphate accumulation and L-citrulline conversion from L-arginine. Nuclear run-on experiments were performed to study the transcription rate of nascent NOSIII messenger ribonucleic acid (mRNA). RESULTS Data showed a significant decrease in NOSIII expression after 24-h treatment with glycosylated low-density lipoprotein (glycLDL) and oxidized low-density lipoprotein (oxLDL). Accordingly, we observed a significant dose-dependent reduction in NO bioactivity (p < 0.05 to p < 0.001 vs. untreated cells, native low density lipoprotein [nLDL], glycLDL, and oxLDL). Glyc-oxLDL did not reduce the half-life of NOSIII mRNA or significantly enhance L-citrulline conversion. Nuclear run-on experiments showed that high doses of glyc-oxLDL can reduce the transcription rate of nascent NOSIII mRNA (densitometric analysis revealed a reduction of 25% [p < 0.05 vs. untreated cells, nLDL, and glycLDL] after treatment of cells with 300 microg/ml glyc-oxLDL). The effects of glyc-oxLDL are not related to the higher levels of oxidative compounds in comparison to those of oxLDL. CONCLUSIONS These results indicate that glyc-oxLDL, per se, may influence signal transduction pathways involving NO-mediated regulatory signals and NOSIII activity in human endothelial cells. This phenomenon can adversely influence the evolution of clinical vascular complications, coronary heart disease, and atherogenesis in diabetic patients.
Collapse
Affiliation(s)
- Claudio Napoli
- Department of Medicine-0682, University of California, San Diego, California 92093, USA.
| | | | | | | | | |
Collapse
|
37
|
Li H, Wallerath T, Förstermann U. Physiological mechanisms regulating the expression of endothelial-type NO synthase. Nitric Oxide 2002; 7:132-47. [PMID: 12223183 DOI: 10.1016/s1089-8603(02)00127-1] [Citation(s) in RCA: 163] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Although endothelial nitric oxide synthase (eNOS) is a constitutively expressed enzyme, its expression is regulated by a number of biophysical, biochemical, and hormonal stimuli, both under physiological conditions and in pathology. This review summarizes the recent findings in this field. Shear stress, growth factors (such as transforming growth factor-beta, fibroblast growth factor, vascular endothelial growth factor, and platelet-derived growth factor), hormones (such as estrogens, insulin, angiotensin II, and endothelin 1), and other compounds (such as lysophosphatidylcholine) upregulate eNOS expression. On the other hand, the cytokine tumor necrosis factor-alpha and bacterial lipopolysaccharide downregulate the expression of this enzyme. The growth status of cells, the actin cytoskeleton, and NO itself are also important regulators of eNOS expression. Both transcriptional and posttranscriptional mechanisms are involved in the expressional regulation of eNOS. Different signaling pathways are involved in the regulation of eNOS promoter activity and eNOS mRNA stability. Changes in eNOS expression and activity under pathophysiological conditions and the pharmacological modulation of eNOS expression are subject of a subsequent brief review (part 2) to be published in the next issue of this journal.
Collapse
Affiliation(s)
- Huige Li
- Department of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Strasse 67, Mainz D-55101, Germany
| | | | | |
Collapse
|
38
|
Govers R, Bevers L, de Bree P, Rabelink TJ. Endothelial nitric oxide synthase activity is linked to its presence at cell-cell contacts. Biochem J 2002; 361:193-201. [PMID: 11772391 PMCID: PMC1222299 DOI: 10.1042/0264-6021:3610193] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The enzyme endothelial nitric oxide synthase (eNOS) is essential for vascular integrity. Many studies have demonstrated a link between the localization and activity of eNOS. Here, we studied the influence of cell-cell contact on this link in the microvascular endothelial bEnd.3 cell line. By immunofluorescence microscopy, eNOS localization at the plasma membrane was found to be dependent on cell-cell contact. In particular, eNOS was highly enriched at the intercellular contact sites. Further analysis showed that the pattern of eNOS localization at the plasma membrane resembled that of PECAM-1 (platelet endothelial cell adhesion molecule 1), but not that of the adherens junction proteins VE (vascular endothelial)-cadherin and plakoglobin. eNOS that was localized at the contact sites was, in part, Triton X-100-insoluble, in contrast with eNOS at the Golgi complex, which may indicate an association of eNOS with the actin cytoskeleton. Interestingly, eNOS activity was up-regulated in confluent monolayers compared with subconfluent cells, while there was no difference in eNOS expression. This correlation between cell confluence and eNOS activity was also found when primary bovine aortic endothelial cells were studied. These data imply that cell-cell contact induces the localization of eNOS at intercellular junctions, which is required for agonist-induced eNOS activation.
Collapse
Affiliation(s)
- Roland Govers
- Department of Vascular Medicine, UMC Utrecht, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
39
|
Misquitta CM, Iyer VR, Werstiuk ES, Grover AK. The role of 3'-untranslated region (3'-UTR) mediated mRNA stability in cardiovascular pathophysiology. Mol Cell Biochem 2001; 224:53-67. [PMID: 11693200 DOI: 10.1023/a:1011982932645] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Knowledge of transcription and translation has advanced our understanding of cardiac diseases. Here, we present the hypothesis that the stability of mRNA mediated by the 3'-untranslated region (3'-UTR) plays a role in changing gene expression in cardiovascular pathophysiology. Several proteins that bind to sequences in the 3'-UTR of mRNA of cardiovascular targets have been identified. The affected mRNAs include those encoding beta-adrenergic receptors, angiotensin II receptors, endothelial and inducible nitric oxide synthases, cyclooxygenase, endothelial growth factor, tissue necrosis factor (TNF-alpha), globin, elastin, proteins involved in cell cycle regulation, oncogenes, cytokines and lymphokines. We discuss: (a) the types of 3'-UTR sequences involved in mRNA stability, (b) AUF1, HuR and other proteins that bind to these sequences to either stabilize or destabilize the target mRNAs, and (c) the potential role of the 3'-UTR mediated mRNA stability in heart failure, myocardial infarction and hypertension. We hope that these concepts will aid in better understanding cardiovascular diseases and in developing new therapies.
Collapse
Affiliation(s)
- C M Misquitta
- Department of Biology, McMaster University, Hamilton, ON, Canada
| | | | | | | |
Collapse
|
40
|
Detich N, Ramchandani S, Szyf M. A conserved 3'-untranslated element mediates growth regulation of DNA methyltransferase 1 and inhibits its transforming activity. J Biol Chem 2001; 276:24881-90. [PMID: 11335728 DOI: 10.1074/jbc.m103056200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ectopic expression of DNA methyltransferase 1 (DNMT1) has been proposed to play an important role in cancer. dnmt1 mRNA is undetectable in growth-arrested cells but is induced upon entrance into the S phase of the cell cycle, and until now, the mechanisms responsible for this regulation were unknown. In this report, we demonstrate that the 3'-untranslated region (3'-UTR) of the dnmt1 mRNA can confer a growth-dependent regulation on its own message as well as a heterologous beta-globin mRNA. Our results indicate that a 54-nucleotide highly conserved element within the 3'-UTR is necessary and sufficient to mediate this regulation. Cell-free mRNA decay experiments demonstrate that this element increases mRNA turnover rates and does so to a greater extent in the presence of extracts prepared from arrested cells. A specific RNA-protein complex is formed with the 3'-UTR only in growth-arrested cells, and a UV cross-linking analysis revealed a 40-kDa protein (p40), the binding of which is dramatically increased in growth-arrested cells and is inversely correlated with dnmt1 mRNA levels as cells are induced into the cell cycle. Although ectopic expression of human DNMT1 lacking the 3'-UTR can transform NIH-3T3 cells, inclusion of the 3'-UTR prevents transformation. These results support the hypothesis that deregulated expression of DNMT1 with the cell cycle is important for cellular transformation.
Collapse
Affiliation(s)
- N Detich
- Department of Pharmacology and Therapeutics, McGill University, 3655 Sir William Osler Promenade, Montreal, Quebec H3G 1Y6, Canada
| | | | | |
Collapse
|
41
|
Nickenig G, Michaelsen F, Müller C, Vogel T, Strehlow K, Böhm M. Post-transcriptional regulation of the AT1 receptor mRNA. Identification of the mRNA binding motif and functional characterization. FASEB J 2001; 15:1490-2. [PMID: 11387267 DOI: 10.1096/fj.00-0842fje] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- G Nickenig
- Klinik und Poliklinik Innere Medizin III, Universität des Saarlandes, 66421 Homburg, Germany.
| | | | | | | | | | | |
Collapse
|
42
|
Solhaug MJ, Kullaprawithaya U, Dong XQ, Dong KW. Expression of endothelial nitric oxide synthase in the postnatal developing porcine kidney. Am J Physiol Regul Integr Comp Physiol 2001; 280:R1269-75. [PMID: 11294743 DOI: 10.1152/ajpregu.2001.280.5.r1269] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The postnatal pattern of renal endothelial nitric oxide synthase (eNOS) is unknown. The purpose of this study was to characterize eNOS expression during maturation and compare this to neuronal NOS (nNOS). The experiments measured whole kidney eNOS mRNA expression by RT-PCR and protein content by Western blot, as well as cortical and medullary protein content in piglets at selected postnatal ages and in adult pigs. Whole kidney eNOS mRNA was compared with nNOS. Whole kidney eNOS expression decreased from the newborn to its lowest at 7 days, returning by 14 days to adult levels. This eNOS mRNA pattern contrasted with nNOS, which was highest at birth, and progressively decreased to its lowest level in the adult. At birth, cortical eNOS protein was greater than medullary, contrasting with the adult pattern of equivalent levels. In conclusion eNOS is developmentally regulated during early renal maturation and may critically participate in renal function during this period. The eNOS developmental pattern differs from nNOS, suggesting that these isoforms may have different regulatory factors and functional contributions in the postnatal kidney.
Collapse
Affiliation(s)
- M J Solhaug
- Department of Pediatrics, Eastern Virginia Medical School, Norfolk, Virginia 23501, USA.
| | | | | | | |
Collapse
|
43
|
Affiliation(s)
- W Lewis
- Department of Pathology, Emory University School of Medicine, 7117 Woodruff Memorial Building, 1639 Pierce Drive, Atlanta, GA 30322, USA.
| |
Collapse
|
44
|
Shi Y, Pritchard KA, Holman P, Rafiee P, Griffith OW, Kalyanaraman B, Baker JE. Chronic myocardial hypoxia increases nitric oxide synthase and decreases caveolin-3. Free Radic Biol Med 2000; 29:695-703. [PMID: 11053770 DOI: 10.1016/s0891-5849(00)00364-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nitric oxide synthase (NOS) is believed to play an important role in protecting the myocardium against ischemia. Chronic hypoxia from birth increases NOS activity in the myocardium resulting in enhanced nitric oxide production and increased resistance to ischemia. We examined the effects of chronic hypoxia on NOS gene and protein expression and on NOS protein association with caveolin-3. Rabbits were raised from birth in a normoxic (F(I)O(2) = 0.21) or a hypoxic (F(I)O(2) = 0.12) environment for 9 d, and then the hearts were isolated. Ribonuclease protection assays revealed that chronic hypoxia did not alter NOS transcript levels for NOS1, NOS2, or NOS3. The most abundant transcript was NOS3. Western analysis revealed NOS3 was the only isoform detected. Immunoblots of NOS3 immunoprecipitates showed that chronic hypoxia increases NOS3 protein by 2.0 +/- 0.4-fold and decreases the amount of caveolin-3 that can be coprecipitated with NOS3 by 5.5 +/- 0.9-fold. Immunoblots of normoxic and hypoxic hearts showed that chronic hypoxia decreases the amount of caveolin-3 in heart homogenates by 2. 2 +/- 0.5-fold. These data suggest that a decrease in caveolin-3 plays a role in the mechanisms by which chronic hypoxia increases NOS3 activity in the myocardium.
Collapse
Affiliation(s)
- Y Shi
- Division of Pediatric Surgery, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Genes contribute significantly to interpopulation differences in vascular disease. Endothelial nitric oxide synthase (eNOS)-a key regulator of vascular nitric oxide production-has been investigated extensively to determine the relevance of DNA variants in the eNOS gene and vascular diseases. Variants in the promoter region, introns, and exons have been explored in a large number of populations but findings have been inconsistent. This paper reviews the current status of functional significance for reported sequence variations in the eNOS gene and the relevance of these variants to different forms of vascular diseases.
Collapse
Affiliation(s)
- X L Wang
- Department of Genetics, Southwest Foundation for Biomedical Research, San Antonio, Texas 78227-5301, USA
| | | |
Collapse
|