1
|
Khachigian LM. The MEK-ERK-Egr-1 axis and its regulation in cardiovascular disease. Vascul Pharmacol 2023; 153:107232. [PMID: 37734428 DOI: 10.1016/j.vph.2023.107232] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
Cardiovascular disease (CVD) is the primary cause of morbidity and mortality in the Western world. Multiple molecular and cellular processes underpinning the pathogenesis of CVD are regulated by the zinc finger transcription factor and product of an immediate-early gene, early growth response-1 (Egr-1). Egr-1 regulates multiple pro-inflammatory processes that underpin the manifestation of CVD. The activity of Egr-1 itself is influenced by a range of post-translational modifications including sumoylation, ubiquitination and acetylation. Egr-1 also undergoes phosphorylation by protein kinases, such as extracellular-signal regulated kinase (ERK) which is itself phosphorylated by MEK. This article reviews recent progress on the MEK-ERK-Egr-1 cascade, notably regulation in conjunction with factors and agents such as TET2, TRIB2, MIAT, SphK1, cAMP, teneligliptin, cholinergic drugs, red wine and flavonoids, wogonin, febuxostat, docosahexaenoic acid and AT1R blockade. Such insights should provide new opportunity for therapeutic intervention in CVD.
Collapse
Affiliation(s)
- Levon M Khachigian
- Vascular Biology and Translational Research, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
2
|
Li Y, Wu B, Hossain MJ, Quagliata L, O'Meara C, Wilkins MR, Corley S, Khachigian LM. Flubendazole inhibits PD-1 and suppresses melanoma growth in immunocompetent mice. J Transl Med 2023; 21:467. [PMID: 37452307 PMCID: PMC10349441 DOI: 10.1186/s12967-023-04289-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitor therapy has revolutionized the clinical management of a diverse range of cancer types, including advanced cutaneous melanoma. While immunotherapy targeting the PD-1/PD-L1 system has become standard of care, overall response rates remain unsatisfactory for most patients and there are no approved small molecule inhibitors of the PD-1/PD-L1 system. Flubendazole (FLU) is an anthelmintic that has been used to treat worm infections in humans and animals for decades. METHODS Here we tested the anti-cancer activity of systemically delivered FLU with suppression of PD-1 in immunocompetent mice. RESULTS In C57BL/6J mice bearing subcutaneous B16F10 melanoma, FLU reduced both tumor growth and PD-1 protein levels without affecting levels of PD-L1. FLU's suppression of PD-1 was accompanied by increased CD3+ T cell infiltration. Western blotting with extracts from human Jurkat T cells showed that FLU inhibited PD-1 protein expression, findings confirmed by flow cytometry. To gain mechanistic insights on FLU's ability to suppress PD-1 protein levels, we performed bulk RNA sequencing on extracts of Jurkat T cells exposed to the benzimidazole for 4 h. From a pool of 14,475 genes there were 1218 differentially-expressed genes; 687 with increased expression and 531 with decreased expression. Among the genes induced by FLU was the AP-1 family member, JUN and surprisingly, pdcd1. KEGG pathway analysis showed FLU up-regulated genes over-represented in multiple pathways (p < 0.01), the top hit being amoebiasis. FLU also affected the expression of genes in cancer-associated pathways, both through down-regulation and up-regulation. Gene set enrichment analysis revealed a large number of immunological signature gene sets correlated with FLU treatment, including gene sets associated with T cell differentiation, proliferation and function. The AP-1 inhibitor T5224 rescued PD-1 protein expression from inhibition by FLU. CONCLUSION This study is the first to show that FLU can inhibit melanoma growth with PD-1 suppression in immunocompetent mice.
Collapse
Affiliation(s)
- Yue Li
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Ben Wu
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Md Jakir Hossain
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Lily Quagliata
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Connor O'Meara
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
- Department of Otorhinolaryngology, Head & Neck Surgery, Prince of Wales Hospital, Randwick, NSW, 2031, Australia
| | - Marc R Wilkins
- Systems Biology Initiative, Ramaciotti Centre for Genomics, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Susan Corley
- Systems Biology Initiative, Ramaciotti Centre for Genomics, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
3
|
Khachigian LM, Black BL, Ferdinandy P, De Caterina R, Madonna R, Geng YJ. Transcriptional regulation of vascular smooth muscle cell proliferation, differentiation and senescence: Novel targets for therapy. Vascul Pharmacol 2022; 146:107091. [PMID: 35896140 DOI: 10.1016/j.vph.2022.107091] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 10/16/2022]
Abstract
Vascular smooth muscle cells (SMC) possess a unique cytoplasticity, regulated by transcriptional, translational and phenotypic transformation in response to a diverse range of extrinsic and intrinsic pathogenic factors. The mature, differentiated SMC phenotype is physiologically typified transcriptionally by expression of genes encoding "contractile" proteins, such as SMα-actin (ACTA2), SM-MHC (myosin-11) and SM22α (transgelin). When exposed to various pathological conditions (e.g., pro-atherogenic risk factors, hypertension), SMC undergo phenotypic modulation, a bioprocess enabling SMC to de-differentiate in immature stages or trans-differentiate into other cell phenotypes. As recent studies suggest, the process of SMC phenotypic transformation involves five distinct states characterized by different patterns of cell growth, differentiation, migration, matrix protein expression and declined contractility. These changes are mediated via the action of several transcriptional regulators, including myocardin and serum response factor. Conversely, other factors, including Kruppel-like factor 4 and nuclear factor-κB, can inhibit SMC differentiation and growth arrest, while factors such as yin yang-1, can promote SMC differentiation whilst inhibiting proliferation. This article reviews recent advances in our understanding of regulatory mechanisms governing SMC phenotypic modulation. We propose the concept that transcription factors mediating this switching are important biomarkers and potential pharmacological targets for therapeutic intervention in cardiovascular disease.
Collapse
Affiliation(s)
- Levon M Khachigian
- Vascular Biology and Translational Research, Department of Pathology, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Brian L Black
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States of America
| | - Péter Ferdinandy
- Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| | - Raffaele De Caterina
- Cardiovascular Division, Pisa University Hospital & University of Pisa, Via Paradisa, 2, Pisa 56124, Italy
| | - Rosalinda Madonna
- Cardiovascular Division, Pisa University Hospital & University of Pisa, Via Paradisa, 2, Pisa 56124, Italy; Division of Cardiovascular Medicine, Department of Internal Medicine, The Center for Cardiovascular Biology and Atherosclerosis Research, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Yong-Jian Geng
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Center for Cardiovascular Biology and Atherosclerosis Research, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| |
Collapse
|
4
|
Santiago FS, Li Y, Khachigian LM. Serine 26 in Early Growth Response-1 Is Critical for Endothelial Proliferation, Migration, and Network Formation. J Am Heart Assoc 2021; 10:e020521. [PMID: 34476983 PMCID: PMC8649526 DOI: 10.1161/jaha.120.020521] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Vascular endothelial cell proliferation, migration, and network formation are key proangiogenic processes involving the prototypic immediate early gene product, Egr‐1 (early growth response‐1). Egr‐1 undergoes phosphorylation at a conserved Ser26 but its function is completely unknown in endothelial cells or any other cell type. Methods and Results A CRISPR/Cas9 strategy was used to introduce a homozygous Ser26>Ala mutation into endogenous Egr‐1 in human microvascular endothelial cells. In the course of generating mutant cells, we produced cells with homozygous deletion in Egr‐1 caused by frameshift and premature termination. We found that Ser26 mutation in Egr‐1, or Egr‐1 deletion, perturbed endothelial cell proliferation in models of cell counting or real‐time growth using the xCELLigence System. We found that Ser26 mutation or Egr‐1 deletion ameliorated endothelial cell migration toward VEGF‐A165 (vascular endothelial growth factor‐A) in a dual‐chamber model. On solubilized basement membrane preparations, Ser26 mutation or Egr‐1 deletion prevented endothelial network (or tubule) formation, an in vitro model of angiogenesis. Flow cytometry further revealed that Ser26 mutation or Egr‐1 deletion elevated early and late apoptosis. Finally, we demonstrated that Ser26 mutation or Egr‐1 deletion increased VE‐cadherin (vascular endothelial cadherin) expression, a regulator of endothelial adhesion and signaling, permeability, and angiogenesis. Conclusions These findings not only indicate that Egr‐1 is essential for endothelial cell proliferation, migration, and network formation, but also show that point mutation in Ser26 is sufficient to impair each of these processes and trigger apoptosis as effectively as the absence of Egr‐1. This highlights the importance of Ser26 in Egr‐1 for a range of proangiogenic processes.
Collapse
Affiliation(s)
- Fernando S Santiago
- Vascular Biology and Translational Research School of Medical Sciences UNSW Medicine and HealthUniversity of New South Wales Sydney NSW Australia
| | - Yue Li
- Vascular Biology and Translational Research School of Medical Sciences UNSW Medicine and HealthUniversity of New South Wales Sydney NSW Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research School of Medical Sciences UNSW Medicine and HealthUniversity of New South Wales Sydney NSW Australia
| |
Collapse
|
5
|
Xu T, Fan X, Zhao M, Wu M, Li H, Ji B, Zhu X, Li L, Ding H, Sun M, Xu Z, Gao Q. DNA Methylation-Reprogrammed Ang II (Angiotensin II) Type 1 Receptor-Early Growth Response Gene 1-Protein Kinase C ε Axis Underlies Vascular Hypercontractility in Antenatal Hypoxic Offspring. Hypertension 2020; 77:491-506. [PMID: 33342239 DOI: 10.1161/hypertensionaha.120.16247] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
As the most common clinical stress during mid and late pregnancy, antenatal hypoxia has profound adverse effects on individual's vascular health later in life, but the underlying mechanisms are still not understood. The purpose of this study was to reveal the mechanisms of the acquired vascular dysfunction in offspring imposed by antenatal hypoxia. Pregnant rats were housed in a normoxic or hypoxic (10.5% oxygen) chamber from gestation day 10 to 21. Male offspring were euthanized at gestational day 21 (fetus) or postnatal 16 weeks old (adult offspring). Mesenteric arteries were collected for examining Ang II (angiotensin II)-mediated vascular contractility, gene expression, and promoter methylation. Antenatal hypoxia increased vascular sensitivity to Ang II, which was resulted by an upregulated AT1R (angiotensin II type 1 receptor). The increased AT1R was correlated with a hypomethylation-mediated activated transcription of Agtr1a (alpha subtype of AT1R). In addition, we presented evidences that there was an AT1R-Egr1 (early growth response gene 1)-PKCε (ε isoform of protein kinase C) axis in vasculature; AT1R could modulate PKCε expression via upregulating Egr1; Egr1 mediated transcription activation of PKCε via Egr1 binding sites in PKCε gene promoter. Overall, antenatal hypoxia activated AT1R-Egr1-PKCε axis in vasculature, eventually predisposed offspring to vascular hypercontractility. This is the first description that antenatal hypoxia resulted in vascular adverse outcomes in postnatal offspring, was strongly associated with reprogrammed gene expression via a DNA methylation-mediated epigenetic mechanism, advancing understanding toward the influence of adverse antenatal factors in early life on long-term vascular health.
Collapse
Affiliation(s)
- Ting Xu
- From the Institute for Fetology (T.X., M.Z., H.L., B.J., X.Z., L.L., M.S., Z.X., Q.G.), First Hospital of Soochow University, Suzhou, China
| | - Xiaorong Fan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.F.)
| | - Meng Zhao
- From the Institute for Fetology (T.X., M.Z., H.L., B.J., X.Z., L.L., M.S., Z.X., Q.G.), First Hospital of Soochow University, Suzhou, China
| | - Meng Wu
- Institutes of Biological and Medical Sciences, Soochow University Medical School, Suzhou, China (M.W.)
| | - Huan Li
- From the Institute for Fetology (T.X., M.Z., H.L., B.J., X.Z., L.L., M.S., Z.X., Q.G.), First Hospital of Soochow University, Suzhou, China
| | - Bingyu Ji
- From the Institute for Fetology (T.X., M.Z., H.L., B.J., X.Z., L.L., M.S., Z.X., Q.G.), First Hospital of Soochow University, Suzhou, China
| | - Xiaolin Zhu
- From the Institute for Fetology (T.X., M.Z., H.L., B.J., X.Z., L.L., M.S., Z.X., Q.G.), First Hospital of Soochow University, Suzhou, China
| | - Lingjun Li
- From the Institute for Fetology (T.X., M.Z., H.L., B.J., X.Z., L.L., M.S., Z.X., Q.G.), First Hospital of Soochow University, Suzhou, China
| | - Hongmei Ding
- Department of Obstetrics and Gynecology (H.D.), First Hospital of Soochow University, Suzhou, China
| | - Miao Sun
- From the Institute for Fetology (T.X., M.Z., H.L., B.J., X.Z., L.L., M.S., Z.X., Q.G.), First Hospital of Soochow University, Suzhou, China
| | - Zhice Xu
- From the Institute for Fetology (T.X., M.Z., H.L., B.J., X.Z., L.L., M.S., Z.X., Q.G.), First Hospital of Soochow University, Suzhou, China
| | - Qinqin Gao
- From the Institute for Fetology (T.X., M.Z., H.L., B.J., X.Z., L.L., M.S., Z.X., Q.G.), First Hospital of Soochow University, Suzhou, China
| |
Collapse
|
6
|
Santiago FS, Sanchez-Guerrero E, Zhang G, Zhong L, Raftery MJ, Khachigian LM. Extracellular signal-regulated kinase-1 phosphorylates early growth response-1 at serine 26. Biochem Biophys Res Commun 2019; 510:345-351. [PMID: 30711252 DOI: 10.1016/j.bbrc.2019.01.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 01/04/2019] [Indexed: 11/30/2022]
Abstract
Egr-1, an immediate-early gene product and master regulator was originally described as a phosphoprotein following its discovery in the 1980s. However specific residue(s) phosphorylated in Egr-1 remain elusive. Here we phosphorylated recombinant Egr-1 in vitro with ERK1 prior to mass spectrometry, which identified phosphorylation of Ser12 and Ser26 with the latter ∼12 times more abundant than Ser12. Phosphorylation of wild-type recombinant Egr-1 (as compared with Ser26>Ala26 mutant Egr-1) revealed that Ser26 accounts for the majority of phosphorylation of Egr-1 by ERK1. N-FGSFPH(pS)PTMDNYC-C was used as an antigen to generate mouse monoclonal antibodies (pS26 MAb). pS26 MAb recognised ERK1-phosphorylated Egr-1 but not Egr-1 bearing a point mutation at Ser26. pS26 MAb recognised inducible ∼75 kDa and 100 kDa species in nuclear extracts of cells exposed to FGF-2. Peptide blocking revealed both inducible species were phosphosite-specific. Immunoprecipitation of nuclear extracts of cells exposed to FGF-2 with pS26 MAb followed by SDS-PAGE and mass spectrometry identified Egr-1 sequences corresponding to the ∼75 kDa species but not ∼100 kDa species. This study identifies a specific amino acid phosphorylated in endogenous Egr-1.
Collapse
Affiliation(s)
- Fernando S Santiago
- Vascular Biology and Translational Research Laboratory, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | | | - Guishui Zhang
- UNSW Medicine, University of New South Wales, Sydney, Australia
| | - Ling Zhong
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, Australia
| | - Mark J Raftery
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research Laboratory, School of Medical Sciences, University of New South Wales, Sydney, Australia; UNSW Medicine, University of New South Wales, Sydney, Australia.
| |
Collapse
|
7
|
Yamashiro Y, Thang BQ, Shin SJ, Lino CA, Nakamura T, Kim J, Sugiyama K, Tokunaga C, Sakamoto H, Osaka M, Davis EC, Wagenseil JE, Hiramatsu Y, Yanagisawa H. Role of Thrombospondin-1 in Mechanotransduction and Development of Thoracic Aortic Aneurysm in Mouse and Humans. Circ Res 2018; 123:660-672. [PMID: 30355232 PMCID: PMC6211815 DOI: 10.1161/circresaha.118.313105] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023]
Abstract
RATIONALE Abnormal mechanosensing of smooth muscle cells (SMCs) resulting from the defective elastin-contractile units has been suggested to drive the formation of thoracic aortic aneurysms; however, the precise molecular mechanism has not been elucidated. OBJECTIVE The aim of this study was to identify the crucial mediator(s) involved in abnormal mechanosensing and propagation of biochemical signals during the aneurysm formation and to establish a basis for a novel therapeutic strategy. METHODS AND RESULTS We used a mouse model of postnatal ascending aortic aneurysms ( Fbln4SMKO; termed SMKO [SMC-specific knockout]), in which deletion of Fbln4 (fibulin-4) leads to disruption of the elastin-contractile units caused by a loss of elastic lamina-SMC connections. In this mouse, upregulation of Egr1 (early growth response 1) and angiotensin-converting enzyme leads to activation of Ang II (angiotensin II) signaling. Here, we showed that the matricellular protein, Thbs1 (thrombospondin-1), was highly upregulated in SMKO ascending aortas and in human thoracic aortic aneurysms. Thbs1 was induced by mechanical stretch and Ang II in SMCs, for which Egr1 was required, and reduction of Fbln4 sensitized the cells to these stimuli and led to higher expression of Egr1 and Thbs1. Deletion of Thbs1 in SMKO mice prevented the aneurysm formation in ≈80% of DKO (SMKO;Thbs1 knockout) animals and suppressed Ssh1 (slingshot-1) and cofilin dephosphorylation, leading to the formation of normal actin filaments. Furthermore, elastic lamina-SMC connections were restored in DKO aortas, and mechanical testing showed that structural and material properties of DKO aortas were markedly improved. CONCLUSIONS Thbs1 is a critical component of mechanotransduction, as well as a modulator of elastic fiber organization. Maladaptive upregulation of Thbs1 results in disruption of elastin-contractile units and dysregulation of actin cytoskeletal remodeling, contributing to the development of ascending aortic aneurysms in vivo. Thbs1 may serve as a potential therapeutic target for treating thoracic aortic aneurysms.
Collapse
MESH Headings
- Actin Cytoskeleton/metabolism
- Actin Cytoskeleton/pathology
- Aged
- Aged, 80 and over
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/prevention & control
- Cells, Cultured
- Cofilin 2/metabolism
- Dilatation, Pathologic
- Disease Models, Animal
- Early Growth Response Protein 1/metabolism
- Elastic Tissue/metabolism
- Elastic Tissue/pathology
- Elastin/metabolism
- Extracellular Matrix Proteins/deficiency
- Extracellular Matrix Proteins/genetics
- Female
- Humans
- Male
- Mechanotransduction, Cellular
- Mice, Knockout
- Middle Aged
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Phosphoprotein Phosphatases/metabolism
- Phosphorylation
- Pressoreceptors/metabolism
- Rats
- Stress, Mechanical
- Thrombospondin 1/deficiency
- Thrombospondin 1/genetics
- Thrombospondin 1/metabolism
- Vascular Remodeling
Collapse
Affiliation(s)
- Yoshito Yamashiro
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Bui Quoc Thang
- Cardiovascular Surgery, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Seung Jae Shin
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki 305-8577, Japan
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Caroline Antunes Lino
- Anatomy, University of Sao Paulo, Institute of Biomedical Sciences, Sao Paulo, SP 05508-900, Brazil
| | | | - Jungsil Kim
- Mechanical Engineering and Materials Science, Washington University, St. Louis, MO 63130, USA
| | - Kaori Sugiyama
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki 305-8577, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Chiho Tokunaga
- Cardiovascular Surgery, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Hiroaki Sakamoto
- Cardiovascular Surgery, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Motoo Osaka
- Cardiovascular Surgery, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Elaine C. Davis
- Anatomy and Cell Biology, McGill University, Montreal, Quebec H3A0C7, Canada
| | - Jessica E. Wagenseil
- Mechanical Engineering and Materials Science, Washington University, St. Louis, MO 63130, USA
| | - Yuji Hiramatsu
- Cardiovascular Surgery, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki 305-8577, Japan
- Faculty of Medicine, University of Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
8
|
Abstract
The renin-angiotensin system (RAS) is a key regulator of blood pressure and blood volume homeostasis. The RAS is primarily comprised of the precursor protein angiotensinogen and the two proteases, renin and angiotensin-converting enzyme (ACE). Angiotensin I (Ang I) is derived from angiotensinogen by renin, but appears to have no biological activity. In contrast, angiotensin II (Ang II) that has a variety of biological functions in the cells is converted from Ang I through removal of two-C-terminal residues by ACE. The physiological effects of Ang II are due to Ang II signaling through specific receptor binding, resulting in muscle contraction leading to increased blood pressure and volume. To modulate RAS, three classes of drugs have been developed: (1) renin inhibitors to prevent angiotensinogen conversion to Ang I, (2) ACE inhibitors, to prevent Ang I processing to Ang II and (3) angiotensin receptor blockers, to inhibit Ang II signaling through its receptor. Studies using the RAS inhibitors and Ang II demonstrated that RAS signaling mediates actions of Ang II in the regulation of proliferation and differentiation of specific hematopoietic cell types, especially in the red blood cell lineage. Accumulating evidence indicates that RAS regulates EPO, an essential mediator of red cell production, for human anemia and erythropoiesis in vivo and in vitro. The regulation of EPO expression by Ang II may be responsible for maintaining red blood cell homeostasis. This review highlights the biological roles of RAS for blood cell and EPO homeostasis through Ang II signaling. The molecular mechanism for Ang II-induced EPO production of the cell or tissue type-specific expression is discussed.
Collapse
Affiliation(s)
- Yong-Chul Kim
- Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Ognoon Mungunsukh
- Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Regina M Day
- Uniformed Services University of the Health Sciences, Bethesda, MD, United States.
| |
Collapse
|
9
|
Simo-Cheyou ER, Vardatsikos G, Srivastava AK. Src tyrosine kinase mediates endothelin-1-induced early growth response protein-1 expression via MAP kinase-dependent pathways in vascular smooth muscle cells. Int J Mol Med 2016; 38:1879-1886. [PMID: 27748819 DOI: 10.3892/ijmm.2016.2767] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 10/04/2016] [Indexed: 11/06/2022] Open
Abstract
We have previously demonstrated that the non-receptor protein tyrosine kinase (NR-PTK) c-Src is an upstream regulator of endothelin-1 (ET-1) and angiotensin II-induced activation of protein kinase B (PKB) signaling in vascular smooth muscle cells (VSMCs). We have also demonstrated that ET-1 potently induces the expression of the early growth response protein-1 (Egr-1), a zinc finger transcription factor that is overexpressed in models of vascular diseases, such as atherosclerosis. However, the involvement of c-Src in ET-1‑induced Egr-1 expression has not yet been investigated and its role in mitogen-activated protein kinase (MAPK) signaling remains controversial. Therefore, the aim of the present study was to examine the role of c-Src in the ET-1-induced phosphorylation of extracellular signal-regulated kinase (ERK)1/2, c-Jun N-terminal kinase (JNK) and p38 MAPK, 3 key members of the MAPK family and in the regulation of Egr-1 expression in rat aortic A10 VSMCs. ET-1 rapidly induced the phosphorylation of MAPKs, as well as the expression of Egr-1; however, treatment of the VSMCs with PP2, a specific pharmacological inhibitor of c-Src, dose-dependently reduced the phosphorylation of the 3 MAPKs and the expression of Egr-1 induced by ET-1. Furthermore, in mouse embryonic fibroblasts (MEFs) deficient in c-Src (SYF), the ET-1-induced Egr-1 expression and MAPK phosphorylation were significantly suppressed, as compared to MEFs expressing normal Src levels. These results suggest that c-Src plays a critical role in mediating ET-1-induced MAPK phosphorylation and Egr-1 expression in VSMCs.
Collapse
Affiliation(s)
- Estelle R Simo-Cheyou
- Laboratory of Cellular Signaling, Montreal Diabetes Research Center, Research Center - University of Montreal Hospital Center, Montreal, QC H2X 0A9, Canada
| | - George Vardatsikos
- Laboratory of Cellular Signaling, Montreal Diabetes Research Center, Research Center - University of Montreal Hospital Center, Montreal, QC H2X 0A9, Canada
| | - Ashok K Srivastava
- Laboratory of Cellular Signaling, Montreal Diabetes Research Center, Research Center - University of Montreal Hospital Center, Montreal, QC H2X 0A9, Canada
| |
Collapse
|
10
|
Ha YM, Nam JO, Kang YJ. Pitavastatin Regulates Ang II Induced Proliferation and Migration via IGFBP-5 in VSMC. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 19:499-506. [PMID: 26557016 PMCID: PMC4637352 DOI: 10.4196/kjpp.2015.19.6.499] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 07/28/2015] [Accepted: 08/16/2015] [Indexed: 11/15/2022]
Abstract
Angiotensin II (Ang II), a key mediator of hypertensive, causes structural changes in the arteries (vascular remodeling), which involve alterations in cell growth, vascular smooth muscle cell (VSMC) hypertrophy. Ang II promotes fibrotic factor like IGFBP5, which mediates the profibrotic effects of Ang II in the heart and kidneys, lung and so on. The purpose of this study was to identify the signaling pathway of IGFBP5 on cell proliferation and migration of Ang II-stimulated VSMC. We have been interested in Ang II-induced IGFBP5 and were curious to determine whether a Pitavastatin would ameliorate the effects. Herein, we investigated the question of whether Ang II induced the levels of IGFBP5 protein followed by proliferation and migration in VSMC. Pretreatment with the specific Angiotensin receptor type 1 (AT1) inhibitor (Losartan), Angiotensin receptor type 2 (AT2) inhibitor (PD123319), MAPK inhibitor (U0126), ERK1/2 inhibitor (PD98059), P38 inhibitor (SB600125) and PI3K inhibitor (LY294002) resulted in significantly inhibited IGFBP5 production, proliferation, and migration in Ang II-stimulated VSMC. In addition, IGFBP5 knockdown resulted in modulation of Ang II induced proliferation and migration via IGFBP5 induction. In addition, Pitavastatin modulated Ang II induced proliferation and migration in VSMC. Taken together, our results indicated that Ang II induces IGFBP5 through AT1, ERK1/2, P38, and PI3K signaling pathways, which were inhibited by Pitavastatin. These findings may suggest that Pitavastatin has an effect on vascular disease including hypertension.
Collapse
Affiliation(s)
- Yu Mi Ha
- Department of Pharmacology, College of Medicine, Yeungnam University, Daegu 42415, Korea
| | - Ju-Ock Nam
- School of Food Science & Biotechnology, Kyungpook National University, Daegu 41566, Korea
| | - Young Jin Kang
- Department of Pharmacology, College of Medicine, Yeungnam University, Daegu 42415, Korea
| |
Collapse
|
11
|
Kim YC, Mungunsukh O, McCart EA, Roehrich PJ, Yee DK, Day RM. Mechanism of erythropoietin regulation by angiotensin II. Mol Pharmacol 2014; 85:898-908. [PMID: 24695083 DOI: 10.1124/mol.113.091157] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Erythropoietin (EPO) is the primary regulator of red blood cell development. Although hypoxic regulation of EPO has been extensively studied, the mechanism(s) for basal regulation of EPO are not well understood. In vivo studies in healthy human volunteers and animal models indicated that angiotensin II (Ang II) and angiotensin converting enzyme inhibitors regulated blood EPO levels. In the current study, we found that Ang II induced EPO expression in situ in murine kidney slices and in 786-O kidney cells in culture as determined by reverse transcription polymerase chain reaction. We further investigated the signaling mechanism of Ang II regulation of EPO in 786-O cells. Pharmacological inhibitors of Ang II type 1 receptor (AT1R) and extracellular signal-regulated kinase 1/2 (ERK1/2) suppressed Ang II transcriptional activation of EPO. Inhibitors of AT2R or Src homology 2 domain-containing tyrosine phosphatase had no effect. Coimmunoprecipiation experiments demonstrated that p21Ras was constitutively bound to the AT1R; this association was increased by Ang II but was reduced by the AT1R inhibitor telmisartan. Transmembrane domain (TM) 2 of AT1R is important for G protein-dependent ERK1/2 activation, and mutant D74E in TM2 blocked Ang II activation of ERK1/2. Ang II signaling induced the nuclear translocation of the Egr-1 transcription factor, and overexpression of dominant-negative Egr-1 blocked EPO promoter activation by Ang II. These data identify a novel pathway for basal regulation of EPO via AT1R-mediated Egr-1 activation by p21Ras-mitogen-activated protein kinase/ERK kinase-ERK1/2. Our current data suggest that Ang II, in addition to regulating blood volume and pressure, may be a master regulator of erythropoiesis.
Collapse
Affiliation(s)
- Yong-Chul Kim
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland (Y.-C.K., O.M., E.A.M., P.J.R., R.M.D.); and Department of Animal Biology, University of Pennsylvania, Philadelphia, Pennsylvania (D.K.Y.)
| | | | | | | | | | | |
Collapse
|
12
|
A repressor protein, Mnt, is a novel negative regulator of vascular smooth muscle cell hypertrophy by angiotensin II and neointimal hyperplasia by arterial injury. Atherosclerosis 2013; 228:90-3. [PMID: 23535568 DOI: 10.1016/j.atherosclerosis.2013.02.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 02/08/2013] [Accepted: 02/25/2013] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The Max-interacting protein Mnt is a transcriptional repressor that can antagonize the transcriptional and proliferation-related activities of Myc. Here, we tested the hypothesis that Mnt is a negative regulator of pathological vascular remodeling. METHODS Adenovirus encoding Mnt or control GFP was infected to cultured rat vascular smooth muscle cells (VSMC) and carotid arteries after a balloon angioplasty. RESULTS In VSMC, adenoviral gene transfer of Mnt suppressed angiotensin II-induced protein expression of early growth response protein-1 (Egr1) and its promoter activation. Mnt adenovirus did not interfere with upstream signaling of angiotensin II. Angiotensin II-induced protein accumulation in VSMC was inhibited by Mnt adenovirus. Mnt adenovirus also inhibited platelet-derived growth factor-induced VSMC proliferation. Moreover, Mnt adenovirus prevented neointima formation in response to arterial injury. The adenoviral Mnt gene transfer also prevented Egr1 induction in neointima. CONCLUSION These data identify Mnt as a previously unrecognized negative regulator of pathological vascular remodeling.
Collapse
|
13
|
Kapakos G, Youreva V, Srivastava AK. Attenuation of endothelin-1-induced PKB and ERK1/2 signaling, as well as Egr-1 expression, by curcumin in A-10 vascular smooth muscle cells. Can J Physiol Pharmacol 2012; 90:1277-85. [DOI: 10.1139/y2012-059] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Endothelin-1 (ET-1) is implicated in the pathogenesis of vascular abnormalities through the hyperactivation of growth promoting pathways, including protein kinase B (PKB) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling. ET-1 has been shown to elicit its responses through the generation of reactive oxygen species (ROS). Curcumin, the main constituent of the spice turmeric, exhibits cardio-protective, anti-proliferative, and antioxidant properties; however, the precise molecular mechanism of its action is unclear. Therefore, in the present study, we investigated the effects of curcumin on ET-1-induced PKB and ERK1/2 signaling, as well as insulin-like growth factor type receptor (IGF-1R) phosphorylation. Curcumin dose-dependently inhibited ET-1-induced phosphorylation of PKB, ERK1/2, c-Raf, and insulin-like growth factor type 1 receptor (IGF-1R), in vascular smooth muscle cells (VSMC). Furthermore, curcumin also attenuated ET-1-induced expression of early growth response (Egr)-1, a transcription factor downstream of ERK1/2 that plays a regulatory role in several cardiovascular pathological processes. In conclusion, these data demonstrate that curcumin is a potent inhibitor of ET-1-induced mitogenic and proliferative signaling events in VSMC and suggest that the ability of curcumin to attenuate these events may contribute as a potential mechanism for its cardiovascular protective response.
Collapse
Affiliation(s)
- Georgia Kapakos
- Laboratory of Cell Signaling, Montreal Diabetes Research Center (MDRC), Research Center of Centre Hospitalier de l’Université de Montréal (CRCHUM) – Angus Campus, Department of Medicine, Université de Montréal, 2901 Rachel Est, Montréal, QC H1W 4A4, Canada
| | - Viktoria Youreva
- Laboratory of Cell Signaling, Montreal Diabetes Research Center (MDRC), Research Center of Centre Hospitalier de l’Université de Montréal (CRCHUM) – Angus Campus, Department of Medicine, Université de Montréal, 2901 Rachel Est, Montréal, QC H1W 4A4, Canada
| | - Ashok K. Srivastava
- Laboratory of Cell Signaling, Montreal Diabetes Research Center (MDRC), Research Center of Centre Hospitalier de l’Université de Montréal (CRCHUM) – Angus Campus, Department of Medicine, Université de Montréal, 2901 Rachel Est, Montréal, QC H1W 4A4, Canada
| |
Collapse
|
14
|
Sanchez-Guerrero E, Chen E, Kockx M, An SW, Chong BH, Khachigian LM. IL-1beta signals through the EGF receptor and activates Egr-1 through MMP-ADAM. PLoS One 2012; 7:e39811. [PMID: 22792188 PMCID: PMC3391205 DOI: 10.1371/journal.pone.0039811] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 05/27/2012] [Indexed: 01/22/2023] Open
Abstract
The immediate-early gene Egr-1 controls the inducible expression of many genes implicated in the pathogenesis of a range of vascular disorders, yet our understanding of the mechanisms controlling the rapid expression of this prototypic zinc finger transcription factor is poor. Here we show that Egr-1 expression induced by IL-1beta is dependent on metalloproteinases (MMP) and a disintegrin and a metalloproteinase (ADAM). Pharmacologic MMP/ADAM inhibitors and siRNA knockdown prevent IL-1beta induction of Egr-1. Further, IL-1beta activates Egr-1 via the epidermal growth factor receptor (EGFR). This is blocked by EGFR tyrosine kinase inhibition and EGFR knockdown. IL-1beta induction of Egr-1 expression is reduced in murine embryonic fibroblasts (mEFs) deficient in ADAM17 despite unbiased expression of EGFR and IL-1RI in ADAM17-deficient and wild-type mEFs. Finally, we show that IL-1beta-inducible wound repair after mechanical injury requires both EGFR and MMP/ADAM. This study reports for the first time that Egr-1 induction by IL-1beta involves EGFR and MMP/ADAM-dependent EGFR phosphorylation.
Collapse
Affiliation(s)
| | - Elya Chen
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - Maaike Kockx
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - Si-Wei An
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - Beng H. Chong
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - Levon M. Khachigian
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
- * E-mail:
| |
Collapse
|
15
|
Takayanagi T, Bourne AM, Kimura K, Takaguri A, Elliott KJ, Eguchi K, Eguchi S. Constitutive stimulation of vascular smooth muscle cells by angiotensin II derived from an adenovirus encoding a furin-cleavable fusion protein. Am J Hypertens 2012; 25:280-3. [PMID: 22113169 DOI: 10.1038/ajh.2011.221] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND To fill the gap between acute and chronic stimulation methods of angiotensin II (Ang II) and obtain relevant signaling information, we have made an adenovirus vector encoding a furin-cleavable Ang II fusion protein. METHODS Vascular smooth muscle cells (VSMCs) were infected with adenovirus to evaluate Ang II production. Also, expression of early growth response-1 (Egr-1) and hypertrophic responses were examined in VSMCs. RESULTS Acute stimulation of VSMCs with synthetic Ang II showed the peptide had a half-life of less than 1 h. Infection of VSMCs with Ang II adenovirus showed a time-dependent production of Ang II as early as 2 days and up to 7 days postinfection. The Ang II adenovirus induced VSMC hypertrophy, stimulated Egr-1 expression, and suppressed Ang II type 1 receptor mRNA expression. Chronic Ang II infusion in mice for 2 weeks markedly enhanced Egr-1 immunostaining in carotid artery compared with the control saline infusion. CONCLUSION Application of the Ang II adenovirus vector to cultured cells will be useful to elucidate molecular and signaling mechanisms of cardiovascular diseases associated with enhanced Ang II production.
Collapse
|
16
|
Rossmann C, Rauh A, Hammer A, Windischhofer W, Zirkl S, Sattler W, Malle E. Hypochlorite-modified high-density lipoprotein promotes induction of HO-1 in endothelial cells via activation of p42/44 MAPK and zinc finger transcription factor Egr-1. Arch Biochem Biophys 2011; 509:16-25. [PMID: 21354100 PMCID: PMC3081070 DOI: 10.1016/j.abb.2011.02.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 02/04/2011] [Accepted: 02/17/2011] [Indexed: 11/15/2022]
Abstract
Modification/chlorination of high-density lipoprotein (HDL) by hypochlorous acid (HOCl), formed by the myeloperoxidase-H₂O₂-chloride system of activated phagocytes, converts an anti-atherogenic lipoprotein into a pro-inflammatory lipoprotein particle. Chlorinated HDL is present in human lesion material, binds to and is internalized by endothelial cells and impairs expression and activity of endothelial nitric oxide synthase. The present study aimed at clarifying whether exposure of endothelial cells to pro-inflammatory HOCl-HDL impacts on expression of heme oxygenase-1, a potential rescue pathway against endothelial dysfunction. Our findings revealed that HDL modified by HOCl, added as reagent or generated enzymatically, induced phosphorylation of p42/44 mitogen-activated protein kinase, expression of transcription factor early growth response-1 (Egr-1) and enhanced expression of heme oxygenase-1 in human endothelial cells. Upregulation of heme oxygenase-1 could be blocked by an inhibitor upstream of p42/44 mitogen-activated protein kinase and/or knockdown of Egr-1 by RNA-interference. Electrophoretic mobility shift assays demonstrated HOCl-HDL-mediated induction of the Egr-1 DNA binding activity. Immunocytochemical and immunoblotting experiments demonstrated HOCl-HDL-induced translocation of Egr-1 to the nucleus. The present study demonstrates a novel compensatory pathway against adverse effects of HOCl-HDL, providing cytoprotection in a number of pathological conditions including cardiovascular disease.
Collapse
Affiliation(s)
- Christine Rossmann
- Center for Molecular Medicine, Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | |
Collapse
|
17
|
Wu X, Cheng J, Li P, Yang M, Qiu S, Liu P, Du J. Mechano-sensitive transcriptional factor Egr-1 regulates insulin-like growth factor-1 receptor expression and contributes to neointima formation in vein grafts. Arterioscler Thromb Vasc Biol 2009; 30:471-6. [PMID: 19965784 DOI: 10.1161/atvbaha.109.184259] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vein grafts in a coronary bypass or a hemodialysis access often develop obliterative growth of the neointima. We previously reported that the mechanical stretch-activated insulin-like growth factor-1 receptor (IGF-1/IGF-1R) pathway plays an important role in this remodeling. However, the transcriptional mechanism(s) regulating IGF-1R expression and neointima formation have not been identified. METHODS AND RESULTS Deletion and site-specific mutagenesis analysis of IGF-1R promoter identified that the minimal mechano-responsive promoter element (-270--130) contains 2 consensus sequences for binding of early growth reponse-1 (Egr-1) transcriptional factor. Mechanical stretch stimulated both Egr-1 mRNA (4.6-fold) and protein (5.2-fold) in vascular smooth muscle cells. Interposition of a vein into an artery increased Egr-1 mRNA (7.8+/-2.6-fold vs sham). In vascular smooth muscle cells isolated from Egr-1 knockout mice, mechanical stretch could not increase IGF-1R, and vascular smooth muscle cells proliferation was decreased by 47% compared to wild-type cells. Importantly, the neointima area was reduced by at least 50%, and the lumen-to-media ratio increased by 55% in vein grafts of Egr-1 knockout mice compared with results of wild-type mice. CONCLUSIONS Egr-1 is a mechano-sensitive transcriptional factor that stimulates IGF-1R transcription, resulting in vascular remodeling of vein grafts.
Collapse
Affiliation(s)
- Xiaoqian Wu
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | | | | | | | | | | | | |
Collapse
|
18
|
Malabanan KP, Kanellakis P, Bobik A, Khachigian LM. Activation Transcription Factor-4 Induced by Fibroblast Growth Factor-2 Regulates Vascular Endothelial Growth Factor-A Transcription in Vascular Smooth Muscle Cells and Mediates Intimal Thickening in Rat Arteries Following Balloon Injury. Circ Res 2008; 103:378-87. [DOI: 10.1161/circresaha.107.168682] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Activation transcription factor (ATF)-4 is a member of the ATF/CREB family of basic leucine zipper transcription factors that regulates cellular responses to a variety of stresses. The role of ATF-4 in smooth muscle cells of the vessel wall is completely unknown. Here, we show that ATF-4 expression is induced in smooth muscle cells in response to injury, both in vitro using a model of mechanical injury and in the media of balloon-injured rat carotid arteries. We demonstrate that ATF-4 is activated by fibroblast growth factor (FGF)-2, an injury-induced mitogen, through the phosphatidylinositol 3-kinase pathway. Injury also activates vascular endothelial growth factor (VEGF)-A, whose expression is stimulated by ATF-4 overexpression and exposure to FGF-2. FGF-2 induces ATF-4 binding to a recognition element located in the VEGF-A gene at +1767 bp and luciferase reporter gene expression dependent on this site. Moreover, ATF-4 knockdown with small interfering RNA or ATF-4 deficiency ameliorates FGF-2–inducible VEGF-A expression. Intraluminal delivery of ATF-4 small interfering RNA in rat carotid arteries blocks balloon injury–inducible ATF-4 and VEGF-A expression after 4 hours and intimal thickening after 14 days. These findings reveal, for the first time, the induction of ATF-4 by both vascular injury and FGF-2. ATF-4 serves as a conduit for the inducible expression of 1 growth factor by another during the process of intimal thickening.
Collapse
Affiliation(s)
- Kristine P. Malabanan
- From the Centre for Vascular Research (K.P.M., L.M.K.), School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney; and Baker Heart Research Institute (P.K., A.B.), Melbourne, Australia
| | - Peter Kanellakis
- From the Centre for Vascular Research (K.P.M., L.M.K.), School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney; and Baker Heart Research Institute (P.K., A.B.), Melbourne, Australia
| | - Alexander Bobik
- From the Centre for Vascular Research (K.P.M., L.M.K.), School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney; and Baker Heart Research Institute (P.K., A.B.), Melbourne, Australia
| | - Levon M. Khachigian
- From the Centre for Vascular Research (K.P.M., L.M.K.), School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney; and Baker Heart Research Institute (P.K., A.B.), Melbourne, Australia
| |
Collapse
|
19
|
Sanchez-Guerrero E, Midgley VC, Khachigian LM. Angiotensin II induction of PDGF-C expression is mediated by AT1 receptor-dependent Egr-1 transactivation. Nucleic Acids Res 2008; 36:1941-51. [PMID: 18272536 PMCID: PMC2330232 DOI: 10.1093/nar/gkm923] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Platelet-derived growth factors are a family of mitogens and chemoattractants comprising of four ligand genes (A-, B-, C-, D-chains) implicated in many physiologic and pathophysiologic processes, including atherosclerosis, fibrosis and tumorigenesis. Our understanding of the molecular mechanisms, which regulate PDGF-C transcription remains incomplete. Transient transfection analysis, conventional and quantitative real-time PCR revealed the induction of PDGF-C transcription and mRNA expression in smooth muscle cells (SMCs) exposed to the peptide hormone angiotensin (ATII), which induces Egr-1. Occupancy of a G + C-rich element in the proximal region of the PDGF-C promoter was unaffected by ATII. Instead we discovered, using both nuclear extracts and recombinant proteins with EMSA and ChIP analyses, the existence of a second Egr-1-binding element located 500 bp upstream. ATII induction of PDGF-C transcription is mediated by the angiotensin type 1 receptor (AT1R) and Egr-1 activation through this upstream element. DNAzyme ED5 targeting Egr-1 blocked ATII-inducible PDGF-C expression. Moreover, increased PDGF-C expression after exposure to ATII depends upon the differentiation state of the SMCs. This study demonstrates the existence of this novel ATII-AT1R-Egr-1-PDGF-C axis in SMCs of neonatal origin, but not in adult SMCs, where ATII induces Egr-1 but not PDGF-C.
Collapse
Affiliation(s)
- Estella Sanchez-Guerrero
- The Centre for Vascular Research, The University of New South Wales and Department of Haematology, The Prince of Wales Hospital, Sydney, Australia
| | | | | |
Collapse
|
20
|
Ingram JL, Antao-Menezes A, Mangum JB, Lyght O, Lee PJ, Elias JA, Bonner JC. Opposing actions of Stat1 and Stat6 on IL-13-induced up-regulation of early growth response-1 and platelet-derived growth factor ligands in pulmonary fibroblasts. THE JOURNAL OF IMMUNOLOGY 2006; 177:4141-8. [PMID: 16951379 DOI: 10.4049/jimmunol.177.6.4141] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-13 is a key cytokine involved in airway remodeling in asthma. We previously reported that IL-13 stimulated the mitogenesis of lung fibroblasts via platelet-derived growth factor (PDGF)-AA. In this report, we show that IL-13 increases PDGF-A and PDGF-C mRNA levels through a dual intracellular cascade that requires coactivation of Stat6 and Stat1 to impact transcriptional regulation of the early growth response (Egr)-1 gene, which then drives PDGF expression. Increased levels of PDGF-AA and PDGF-CC protein were observed in vivo in the airways of IL-13 transgenic mice. IL-13 up-regulated PDGF-A and PDGF-C mRNA levels in lung fibroblasts isolated from three different background strains of mice. However, IL-13-induced PDGF-A and PDGF-C mRNA levels were significantly reduced in Stat6-deficient (Stat6(-/-)) fibroblasts as compared with wild-type Stat6(+/+) fibroblasts. In contrast, IL-13-induced PDGF-A and PDGF-C mRNAs were enhanced in Stat1(-/-) fibroblasts as compared with Stat1(+/+) fibroblasts. IL-13 did not up-regulate PDGF-A or PDGF-C mRNA levels in Egr-1(-/-) fibroblasts. Moreover, IL-13 did not increase Egr-1 mRNA and protein levels in Stat6(-/-) fibroblasts and yet enhanced Egr-1 mRNA and protein levels in Stat1(-/-) fibroblasts. Our findings support the hypothesis that Stat6 and Stat1 exert stimulatory and inhibitory effects on Egr-1 and PDGF ligand mRNA transcription, respectively. This novel mechanism could aid in identifying molecular targets for the treatment of chronic airway remodeling and fibrosis in asthma.
Collapse
Affiliation(s)
- Jennifer L Ingram
- CIIT Centers for Health Research, Research Triangle Park, Durham, NC 27709, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
The immediate-early gene product and zinc finger transcription factor early growth response (Egr)-1 plays a key master regulatory role in multiple cardiovascular pathological processes. This article reviews the amazing recent evidence implicating Egr-1 in atherosclerosis, intimal thickening after acute vascular injury, ischemic pathology, angiogenesis, allograft rejection, and cardiac hypertrophy.
Collapse
Affiliation(s)
- Levon M Khachigian
- Centre for Vascular Research, Department of Pathology, The University of New South Wales, The Prince of Wales Hospital, Sydney, Australia.
| |
Collapse
|
22
|
|
23
|
de Mestre AM, Rao S, Hornby JR, Soe-Htwe T, Khachigian LM, Hulett MD. Early growth response gene 1 (EGR1) regulates heparanase gene transcription in tumor cells. J Biol Chem 2005; 280:35136-47. [PMID: 16093249 DOI: 10.1074/jbc.m503414200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Heparanase is an endoglycosidase that degrades heparan sulfate chains of heparan sulfate proteoglycans, a key component of extracellular matrix and basement membranes. Studies using heparanase inhibitors and gene silencing have provided evidence to support an important role for heparanase in tumor metastasis and angiogenesis. The expression of heparanase is normally very tightly controlled, however, it is commonly deregulated in tumor cells, which express elevated heparanase activity that correlates with high levels of heparanase mRNA. We recently identified the transcription factor early growth response gene 1, EGR1, as a key regulator of inducible heparanase transcription in T cells. In this study using chromatin immunoprecipitation, we demonstrate for the first time that EGR1 binds to the heparanase gene promoter in vivo. The important question of the role of EGR1 in regulating heparanase transcription in tumor cells was then assessed. Studies were carried out in four epithelial tumor lines of different tissue origin. Functional dissection of the heparanase promoter identified a 280-bp region that was critical for transcription of the heparanase gene. Transactivation studies using an EGR1 expression vector co-transfected with a reporter construct containing the 280-bp region showed EGR1-activated heparanase promoter activity in a dose-dependent manner in prostate or breast adenocarcinoma and colon carcinoma cell lines. In contrast, overexpression of EGR1 resulted in a dose-dependent repression of promoter activity in melanoma cells. Using site-directed mutagenesis the 280-bp region was found to contain two functional EGR1 sites and electrophoretic mobility shift assays showed binding of EGR1 to both of these sites upon activation of tumor cells. Furthermore, the heparanase promoter region containing the EGR1 sites was also inducible in tumor cells and induction corresponded to HPSE expression levels. These studies show that EGR1 regulates heparanase transcription in tumor cells and importantly, can have a repressive or activating role depending on the tumor type.
Collapse
Affiliation(s)
- Amanda M de Mestre
- Cancer and Vascular Biology Group, Division of Immunology and Genetics, The John Curtin School of Medical Research, The Australian National University, Acton ACT 2601
| | | | | | | | | | | |
Collapse
|
24
|
Pedigo NG, Zhang H, Bruno MEC, Kaetzel CS, Dugan AR, Shanehsaz P, Hennigan RF, Xing Z, Koszewski NJ, Kaetzel DM. A 5'-distal enhanceosome in the PDGF-A gene is activated in choriocarcinoma cells via ligand-independent binding of vitamin D receptor and constitutive jun kinase signaling. Oncogene 2005; 24:2654-66. [PMID: 15829977 DOI: 10.1038/sj.onc.1208336] [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] [Indexed: 11/09/2022]
Abstract
Overexpression of platelet-derived growth factor A-chain (PDGF-A) is clearly linked to autocrine and paracrine stimulation of malignant growth in many human cancers. We have shown previously that PDGF-A overexpression in choriocarcinoma, hepatoma and lung carcinoma cell lines is driven by the activity of a 66 bp enhancer element (ACE66) located approximately 7 kb upstream of the PDGF-A transcription start site. In this study, the ACE66 element is shown to be activated in JEG-3 choriocarcinoma cells through synergistic interactions between consensus DNA motifs for binding of vitamin D receptor, AP1 and ELK1. Binding of the vitamin D/retinoid-X receptor (VDR/RXRalpha) heterodimer to the ACE66 element was reconstituted in vitro with recombinant VDR/RXRalpha and with JEG-3 nuclear extract, and was verified in living JEG-3 cells by chromatin immunoprecipitation analysis. Transcriptional activity of the ACE66 element, as well as occupancy of the element by VDR/RXRalpha, was shown to be independent of stimulation with the hormonal VDR ligand, 1,25-dihydroxyvitamin D3. The jun kinase pathway of mitogen-activated protein kinase (MAPK) signaling was shown to activate the ACE66 enhancer, most likely through activation of factors binding to the AP1 element. These results identify a novel mechanism of transcriptional enhancement involving ligand-independent activity of the VDR/RXR heterodimer and MAPK signaling pathways that appears to play an important role in the overexpression of PDGF in many different settings of human malignancy.
Collapse
Affiliation(s)
- Nancy G Pedigo
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Shenoy SK, Lefkowitz RJ. Receptor-specific Ubiquitination of β-Arrestin Directs Assembly and Targeting of Seven-transmembrane Receptor Signalosomes. J Biol Chem 2005; 280:15315-24. [PMID: 15699045 DOI: 10.1074/jbc.m412418200] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Angiotensin II type 1a (AT1a), vasopressin V2, and neurokinin 1 (NK1) receptors are seven-transmembrane receptors (7TMRs) that bind and co-internalize with the multifunctional adaptor protein, beta-arrestin. These receptors also lead to robust and persistent activation of extracellular-signal regulated kinase 1/2 (ERK1/2) localized on endosomes. Recently, the co-trafficking of receptor-beta-arrestin complexes to endosomes was demonstrated to require stable beta-arrestin ubiquitination (Shenoy, S. K., and Lefkowitz, R. J. (2003) J. Biol. Chem. 278, 14498-14506). We now report that lysines at positions 11 and 12 in beta-arrestin2 are specific and required sites for its AngII-mediated sustained ubiquitination. Thus, upon AngII stimulation the mutant beta-arrestin2(K11,12R) is only transiently ubiquitinated, does not form stable endocytic complexes with the AT1aR, and is impaired in scaffolding-activated ERK1/2. Fusion of a ubiquitin moiety in-frame to beta-arrestin2(K11,12R) restores AngII-mediated trafficking and signaling. Wild type beta-arrestin2 and beta-arrestin2(K11R,K12R)-Ub, but not beta-arrestin2(K11R,K12R), prevent nuclear translocation of pERK. These findings imply that sustained beta-arrestin ubiquitination not only directs co-trafficking of receptor-beta-arrestin complexes but also orchestrates the targeting of "7TMR signalosomes" to microcompartments within the cell. Surprisingly, binding of beta-arrestin2(K11R,K12R) to V2R and NK1R is indistinguishable from that of wild type beta-arrestin2. Moreover, ubiquitination patterns and ERK scaffolding of beta-arrestin2(K11,12R) are unimpaired with respect to V2R stimulation. In contrast, a quintuple lysine mutant (beta-arrestin2(K18R,K107R,K108R,K207R,K296R)) is impaired in endosomal trafficking in response to V2R but not AT1aR stimulation. Our findings delineate a novel regulatory mechanism for 7TMR signaling, dictated by the ubiquitination of beta-arrestin on specific lysines that become accessible for modification due to the specific receptor-bound conformational states of beta-arrestin2.
Collapse
Affiliation(s)
- Sudha K Shenoy
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
26
|
Wei H, Ahn S, Barnes WG, Lefkowitz RJ. Stable Interaction between β-Arrestin 2 and Angiotensin Type 1A Receptor Is Required for β-Arrestin 2-mediated Activation of Extracellular Signal-regulated Kinases 1 and 2. J Biol Chem 2004; 279:48255-61. [PMID: 15355986 DOI: 10.1074/jbc.m406205200] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Binding of beta-arrestins to seven-membrane-spanning receptors (7MSRs) not only leads to receptor desensitization and endocytosis but also elicits additional signaling processes. We recently proposed that stimulation of the angiotensin type 1A (AT(1A)) receptor results in independent beta-arrestin 2- and G protein-mediated extracellular signal-regulated kinases 1 and 2 (ERK1/2) activation. Here we utilize two AT(1A) mutant receptors to study these independent pathways, one truncated at residue 324, thus removing all potential carboxyl-terminal phosphorylation sites, and the other bearing four mutations in the serine/threonine-rich clusters in the carboxyl terminus. As assessed by confocal microscopy, the two mutant receptors interacted with beta-arrestin 2-green fluorescent protein with much lower affinity than did the wild-type receptor. In addition, the mutant receptors more robustly stimulated G protein-mediated inositol phosphate production. Approximately one-half of the wild-type AT(1A) receptor-stimulated ERK1/2 activation was via a beta-arrestin 2-dependent pathway (suppressed by beta-arrestin 2 small interfering RNA), whereas the rest was mediated by a G protein-dependent pathway (suppressed by protein kinase C inhibitor). ERK1/2 activation by the mutant receptors was insensitive to beta-arrestin 2 small interfering RNA but was reduced more than 80% by a protein kinase C inhibitor. The biochemical consequences of ERK activation by the G protein and beta-arrestin 2-dependent pathways were also distinct. G-protein-mediated ERK activation enhanced the transcription of early growth response 1, whereas beta-arrestin 2-dependent ERK activation did not. In addition, stimulation of the truncated AT(1A) mutant receptor caused significantly greater early growth response 1 transcription than did the wild-type receptor. These findings demonstrate how the ability of receptors to interact with beta-arrestins determines both the mechanism of ERK activation as well as the physiological consequences of this activation.
Collapse
Affiliation(s)
- Huijun Wei
- Howard Hughes Medical Institute, Departments of Medicine and Biochemistry, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
27
|
Santiago FS, Khachigian LM. Ets-1 stimulates platelet-derived growth factor A-chain gene transcription and vascular smooth muscle cell growth via cooperative interactions with Sp1. Circ Res 2004; 95:479-87. [PMID: 15297375 DOI: 10.1161/01.res.0000141135.36279.67] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The platelet-derived growth factor (PDGF) family of ligands (composed of A-, B-, C-, and D-chains), potent mitogens, and chemoattractants for cells of mesenchymal origin has been implicated in numerous vascular pathologies involving smooth muscle cell (SMC) hyperplasia. Understanding the molecular mechanisms mediating PDGF transcription would provide new insights into strategies to control PDGF-dependent pathophysiologic processes. We demonstrated previously that PDGF-A expression is under the positive regulatory influence of Sp1, Sp3, and Egr-1 and is negatively controlled by GCF2, NF-1(X), and WT-1. In this article, we demonstrate that Ets-1 induces PDGF-A expression in primary rat aortic SMCs at the level of transcription and mRNA expression. Electrophoretic mobility shift, supershift, and mutational analyses revealed a functional role for the (-555)TTCC(-552) motif in the PDGF-A promoter that binds endogenous Ets-1. Chromatin immunoprecipitation analysis showed the interaction of endogenous and exogenous Ets-1 or glutathione S-transferase-tagged Ets-1, bearing only the DNA-binding domain with the authentic PDGF-A promoter. Conversely, dominant-negative mutant of Ets-1 blocked the promoter interaction of endogenous Ets-1. Overexpression of Ets-1 but not the mutant form of Ets-1 activates the PDGF-A promoter cooperatively with Sp1. Sp1, which interacts with Ets-1, failed to induce PDGF-A promoter-dependent expression if the promoter contained a site-specific mutation in this novel Ets-binding site. Small interfering RNA to Ets-1 and Sp1 blocked PDGF-BB- and serum-inducible PDGF-A expression. SMC growth was stimulated by Ets-1 and Sp1 separately and further increased by both factors together. Ets-1-inducible mitogenesis is blocked by antibodies neutralizing PDGF-A and involves activation of the PDGF alpha-receptor, which binds PDGF-A. These findings identify a functional cis-acting element for Ets-1 in the PDGF-A promoter and demonstrate that Sp1 and Ets-1 cooperatively activate PDGF-A transcription in vascular SMCs.
Collapse
Affiliation(s)
- Fernando S Santiago
- Centre for Vascular Research, The University of New South Wales, and the Department of Haematology, The Prince of Wales Hospital, Sydney, Australia
| | | |
Collapse
|
28
|
de Mestre AM, Khachigian LM, Santiago FS, Staykova MA, Hulett MD. Regulation of Inducible Heparanase Gene Transcription in Activated T Cells by Early Growth Response 1. J Biol Chem 2003; 278:50377-85. [PMID: 14522979 DOI: 10.1074/jbc.m310154200] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cleavage of heparan sulfate by the beta-D-endoglucuronidase heparanase (HPSE) is a fundamental event in a number of important physiological processes including inflammation, wound healing, and angiogenesis. HPSE activity has also been directly correlated with pathological conditions such as tumor growth and metastasis and autoimmune disease. The tight regulation of HPSE expression and function is critical to ensure homeostasis of the normal physiological processes to which it contributes and to prevent imbalance toward pathological situations. Little is known about the transcriptional mechanisms that regulate HPSE expression. In this study we have shown human HPSE gene transcription in Jurkat T cells is induced upon activation. Functional analysis of the HPSE promoter has identified a 280-bp region that is highly inducible. Mutation studies together with supershift experiments have identified a 4-bp motif that binds the transcription factor early growth response-1 (Egr1) and is critical in regulating inducible HPSE gene transcription. Furthermore, the overexpression of Egr1 resulted in the enhanced activation of the HPSE promoter. By using MAPK pathway inhibitors, we have also shown that inducible expression of HPSE mRNA and the activity of the 280-bp HPSE promoter element are dependent on the ERK1/2 (MEK1/2) pathway. This pathway is critical for induction of Egr1 expression at both the mRNA and protein level in T cells, an observation that provides further support to Egr1 playing an important role as a key activator of HPSE expression. In addition, HPSE and Egr1 were shown to co-localize by immunohistochemistry to invading mononuclear leukocytes in actively induced experimental autoimmune encephalomyelitis in rats. These findings provide the first insight into the mechanisms controlling inducible transcription of the HPSE gene, and could represent an important lead into understanding how HPSE expression is deregulated in metastatic tumor cells.
Collapse
Affiliation(s)
- Amanda M de Mestre
- Cancer and Vascular Biology Group, John Curtin School of Medical Research, Australian National University, Acton ACT 2601
| | | | | | | | | |
Collapse
|
29
|
Zhang F, Lin M, Abidi P, Thiel G, Liu J. Specific interaction of Egr1 and c/EBPbeta leads to the transcriptional activation of the human low density lipoprotein receptor gene. J Biol Chem 2003; 278:44246-54. [PMID: 12947119 DOI: 10.1074/jbc.m305564200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The sterol-independent regulatory element (SIRE) of the LDL receptor (LDLR) promoter mediates oncostatin M (OM)-induced transcription of the LDLR gene through a cholesterol-independent pathway. Our prior studies have detected specific associations of the zinc finger transcription factor Egr1 with the SIRE sequence in OM-stimulated HepG2 cells. Because the SIRE motif is composed of a c/EBP binding site and a cAMP response element, both of which are quite divergent from the classical GC-rich Egr1 recognition sequences, we hypothesized that Egr1 may regulate LDLR transcription through interacting with members of the c/EBP and CREB families. Here, we show that treating HepG2 cells with OM specifically leads to prominent increases of the levels of c/EBPbeta and Egr1 bound to the LDLR promoter in vivo. In vitro, the binding of Egr1 to the SIRE sequence is weak, but is strikingly enhanced in the presence of HepG2 nuclear extract. Mammalian two-hybrid assays demonstrate that the N-terminal transactivation domain of Egr1 specifically interacts with c/EBPbeta but not with c/EBPalpha or CREB. The OM treatment further enhances this interaction, resulting in a large increase in the Egr1 transactivating activity. The direct protein to protein contact between Egr1 and c/EBPbeta is also demonstrated by co-immunoprecipitation experiments. Furthermore, we show that a mutation of the phosphorylation motif of c/EBPbeta diminished the OM-stimulated interaction of Egr1 and c/EBPbeta. Taken together, we provide strong evidence that Egr1 regulates LDLR transcription via a novel mechanism of protein-protein interaction with c/EBPbeta.
Collapse
Affiliation(s)
- Fang Zhang
- Department of Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, USA
| | | | | | | | | |
Collapse
|
30
|
Abstract
The molecular mechanisms which control the transcription of growth factor genes underlie such diverse biological processes as embryonic development, cellular differentiation and wound healing. Moreover, disruption of these controls is implicated in the development and progression of a wide variety of human diseases, including cancer, atherosclerosis and fibrotic disease. This review highlights progress made in the study of the gene encoding platelet-derived growth factor A-chain (PDGF-A) from the perspective of its normal patterns of expression, as well as possible mechanisms leading to dysregulation and disease. A particular focus has been placed on the identification and characterization of specific DNA elements, DNA-binding proteins and other aspects of transcriptional regulation involved in activation and repression of the human PDGF-A promoter.
Collapse
Affiliation(s)
- David M Kaetzel
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536-0084, USA.
| |
Collapse
|
31
|
Haznedaroglu IC, Oztürk MA. Towards the understanding of the local hematopoietic bone marrow renin-angiotensin system. Int J Biochem Cell Biol 2003; 35:867-80. [PMID: 12676173 DOI: 10.1016/s1357-2725(02)00278-9] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The classical view of the renin-angiotensin system (RAS) as a circulating endocrine system has evolved to organ- and tissue-based systems that perform paracrine/autocrine functions. Angiotensin II (Ang II), the dominant effector peptide of the RAS, regulates cellular growth in a wide variety of tissues in (patho)biological states. In 1996, we hypothesized that there exists a locally active RAS in the bone marrow affecting the growth, production, proliferation and differentiation of hematopoietic cells. Evidences supporting this hypothesis are growing. Ang II, through interacting with Ang II type 1 (AT1) receptor stimulates erythroid differentiation. This stimulatory effect of Ang II on erythropoiesis was completely abolished by a specific AT1 receptor antagonist, losartan. AT1a receptors are present on human CD34(+) hematopoietic stem cells. Ang II increases hematopoietic progenitor cell proliferation and this effect was also blocked by losartan. Angiotensin-converting enzyme (ACE) is involved in enhancing the recruitment of primitive stem cells into S-phase in hematopoietic bone marrow by degrading tetrapeptide AcSDKP. ACE inhibitors modified the circulating hematopoietic progenitors in healthy subjects. RAS may also affect pathological/neoplastic hematopoiesis. Renin has been isolated from leukemic blast cells. Higher bone marrow ACE levels in acute leukemic patients suggested that ACE is produced at higher quantities in the leukemic bone marrow. In this review, the 'State of the Art' of the local bone marrow RAS is summarized. A local RAS in the bone marrow can mediate, in an autocrine/paracrine fashion, some of the principal steps of hematopoietic cell production. To show a causal link between the components of RAS and the other regulatory hematopoietic growth factors is not only an academic curiosity. Elucidation of such a local bone marrow system may offer novel therapeutic approaches in pathologic/neoplastic conditions.
Collapse
Affiliation(s)
- Ibrahim C Haznedaroglu
- Department of Hematology and Internal Medicine, Hacettepe University Medical School, Sihhiye, Ankara TR-06100, Turkey.
| | | |
Collapse
|
32
|
Vandermeersch S, Stefanovic V, Hus-Citharel A, Ardaillou R, Dussaule JC, Chansel D. AT 1 Receptor Expression in Glomeruli from NO-Deficient Rats. ACTA ACUST UNITED AC 2003; 95:e119-28. [PMID: 14646364 DOI: 10.1159/000074328] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2002] [Accepted: 06/26/2003] [Indexed: 11/19/2022]
Abstract
Chronic inhibition of nitric oxide synthase promotes renin-dependent hypertension and renal injury. The present study examines how renal angiotensin II receptors are expressed in this model. N(G)-nitro-L-arginine methyl ester (L-NAME) was given orally to rats for 1 month and was associated or not with captopril during the 4 last days of the administration. 125I-[Sar1, Ile8]-Ang II binding, AT1)mRNA and cytosolic calcium were studied in isolated glomeruli from L-NAME and control rats and in cultured mesangial cells from normal rats. Renal injury was marked in rats receiving L-NAME. Type I angiotensin II (AT1) receptor number and mRNA expression were decreased (p < 0.05) in glomeruli isolated from L-NAME-treated rats compared with controls, unless they received captopril in combination. The low level of AT1 receptor expression was associated with an attenuated rise of cytosolic calcium in response to angiotensin II. Angiotensin-converting enzyme activity in glomeruli and angiotensin II concentration in renal cortex were increased (p < 0.05) in rats receiving L-NAME alone, whereas aminopeptidase A activity was not modified. To better discriminate between the direct and indirect effects of nitric oxide deficiency, rat mesangial cells were exposed or not for 24 h to S-nitroso-N-acetyl penicillamine, a nitric oxide donor. Angiotensin II binding, AT1 mRNA expression and calcium response to angiotensin II were decreased in presence of the nitric oxide donor (p < 0.01). These results suggest that the decrease of AT1 receptor expression after 1 month of L-NAME treatment does not depend on a direct effect of nitric oxide deficiency but results from the high local angiotensin II concentration due to the stimulated angiotensin-converting enzyme activity. They also show that the renin-angiotensin dependence of this model of hypertension does not result from the overexpression of AT1 receptors.
Collapse
|
33
|
Wilder PJ, Bernadt CT, Kim JH, Rizzino A. Stimulation of the murine type II transforming growth factor-beta receptor promoter by the transcription factor Egr-1. Mol Reprod Dev 2002; 63:282-90. [PMID: 12237943 DOI: 10.1002/mrd.10165] [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: 11/06/2022]
Abstract
Previous studies have demonstrated that differentiation of murine embryonal carcinoma (EC) cells leads to the appearance of high affinity receptors for transforming growth factor-beta (TGF-beta). Subsequently, it was demonstrated that differentiation of F9 EC cells leads to increases in the transcription of the type II TGF-beta-receptor gene (TbetaR-II) and leads to significant increases in the steady-state levels of TbetaR-II mRNA. Analysis of the human TbetaR-II promoter in F9-differentiated cells identified several cis-regulatory elements that influence the activity of the promoter, including a CRE/ATF site and a CCAAT box motif. In the work described in this report, we focused on the effect of the transcription factor Egr-1 on the murine TbetaR-II promoter. We have identified an Egr-1 response-element approximately 150 bp upstream of the major transcription start site of the murine TbetaR-II gene. We demonstrate by electrophoretic mobility shift analysis (EMSA) that this cis-regulatory element binds Egr-1, and we demonstrate that disruption of this site eliminates the response to Egr-1. As part of this analysis, we also examined the effect of Egr-1 on human TbetaR-II promoter. In contrast to a previous report, which reported that Egr-1 inhibits expression of human TbetaR-II promoter/reporter gene constructs, we did not observe an inhibitory effect of Egr-1 that was specific for the human TbetaR-II promoter. Taken together, the findings described in this report identify important differences between the human and the murine TbetaR-II promoter, and our findings identify an Egr-1 cis-regulatory element that is capable of stimulating the activity of the murine TbetaR-II promoter.
Collapse
Affiliation(s)
- Phillip J Wilder
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-6805, USA
| | | | | | | |
Collapse
|
34
|
Platelet-derived growth factor receptor-β in myocyte was upregulated by angiotensin II. CHINESE SCIENCE BULLETIN-CHINESE 2002. [DOI: 10.1007/bf02907573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
35
|
Utsugi M, Dobashi K, Koga Y, Masubuchi K, Shimizu Y, Endou K, Nakazawa T, Mori M. Ambroxol inhibits platelet-derived growth factor production in human monocytic cells. Eur J Pharmacol 2002; 436:47-51. [PMID: 11834245 DOI: 10.1016/s0014-2999(01)01609-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Several growth factors, including platelet-derived growth factor (PDGF), have been implicated in the mechanism of lung and airway remodeling. We investigated the effect of ambroxol, trans-4-[(2-amino-3,5-dibromobenzyl) amino] cyclohexanol hydrochloride, on the lipopolysaccharide-induced PDGF production in human monocytic cells, THP-1. Ambroxol inhibited the lipopolysaccharide-induced PDGF-AB production via PDGF-A mRNA expression. Lipopolysaccharide activated p44/42 extracellular signal-regulated kinase (ERK), and ambroxol attenuated the lipopolysaccharide-induced p44/42 ERK activation. Furthermore, mitogen-activated protein kinase kinase (MEK)-1-specific inhibitor, 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one (PD 98059), blocked the lipopolysaccharide-induced p44/42 ERK activation and PDGF production. These findings indicate that ambroxol inhibits the lipopolysaccharide-induced PDGF production due to the suppression of p44/42 ERK activity.
Collapse
Affiliation(s)
- Mitsuyoshi Utsugi
- First Department of Internal Medicine, Gunma University Faculty of Medicine, School of Medicine, 3-39-15 Showa-machi, Gunma, 371-8511, Maebashi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Rafty LA, Santiago FS, Khachigian LM. NF1/X represses PDGF A-chain transcription by interacting with Sp1 and antagonizing Sp1 occupancy of the promoter. EMBO J 2002; 21:334-43. [PMID: 11823426 PMCID: PMC125828 DOI: 10.1093/emboj/21.3.334] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The regulatory mechanisms mediating basal and inducible platelet-derived growth factor (PDGF)-A expression have been the focus of intense recent investigation, but repression of PDGF-A expression is largely unexplored. Here we isolated a nuclear factor that interacts with the proximal region of the PDGF-A promoter using bulk binding assays and chromatography techniques. Peptide mass fingerprint and supershift analysis revealed this DNA-binding protein to be NF1/X. NF1/X repressed PDGF-A promoter-dependent transcription and endogenous mRNA expression, which was reversible by oligonucleotide decoys bearing an NF1/X-binding site. Mutation in the DNA-binding domain of NF1/X abolished its repression of PDGF-A promoter. NF1/X antagonized the activity of a known activator of the PDGF-A chain, Sp1, by inhibiting its occupancy of the proximal PDGF-A promoter. NF1/X physically and specifically interacts with Sp1 via its subtype-specific domain and blocks Sp1 induction of the promoter. NF1/X residues 311-416 mediated NF1/X suppression of basal PDGF-A transcription, whereas residues 243-416 were required for NF1/X repression of Sp1-inducible promoter activity. These findings demonstrate that repression of PDGF-A gene transcription is governed by interplay between NF1/X and Sp1.
Collapse
Affiliation(s)
| | | | - Levon M. Khachigian
- Centre for Thrombosis and Vascular Research, Department of Pathology, The University of New South Wales and Department of Haematology, Prince of Wales Hospital, Sydney, Australia
Corresponding author e-mail:
| |
Collapse
|
37
|
Bahouth SW, Beauchamp MJ, Vu KN. Reciprocal regulation of beta(1)-adrenergic receptor gene transcription by Sp1 and early growth response gene 1: induction of EGR-1 inhibits the expression of the beta(1)-adrenergic receptor gene. Mol Pharmacol 2002; 61:379-90. [PMID: 11809863 DOI: 10.1124/mol.61.2.379] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The beta(1)-adrenergic receptor (beta(1)-AR) plays a key role in regulating heart rate and contractility in response to catecholamines. Our studies have focused on defining the factors that regulate the expression of the beta(1)-AR gene. We determined that a 65-base-pair (bp) region in the beta(1)-AR promoter between bp -394 and bp -330 directs basal transcription. An element located between -377 and -365 can bind Sp1 and Sp3. In Drosophila melanogaster SL2 cells, Sp1 stimulated the expression of the beta(1)-AR promoter, whereas Sp3 was unable to activate transcription. Site-directed mutagenesis indicated that an intact Sp1-binding site is essential for maintaining the activity of the basal promoter. In addition to binding Sp family members, the nucleotides between -381 and -367 can bind the zinc-finger transcription factor Egr-1. The Egr-1 and Sp1 binding sites are partially overlapping and their binding sequence is conserved among mammalian beta(1)-AR genes. The induction of Egr-1 in rat neonatal ventricular myocytes with phorbol-12-myristate-13-acetate or in HeLa S3 cells by regulated expression of Egr-1 in a tetracycline-responsive promoter, suppressed expression from the beta(1)-AR promoter. Overexpression of Sp1 in SK-N-MC cells increased beta(1)-AR mRNA by 2.4-fold, whereas overexpression of Egr-1 reduced beta(1)-AR mRNA by 40%. Coexpression of Egr-1 with Sp1 reduced Sp1-mediated up-regulation of beta(1)-AR mRNA by 60%. Mutagenesis revealed that an intact Sp1-binding site is essential for observing transcriptional repression by Egr-1 and that Egr-1 suppressed the transcription of the beta(1)-AR gene by competing with Sp1 for binding to their overlapping sites. These results reveal a novel physiologically relevant transcriptional mechanism for reciprocal regulation of beta(1)-AR gene expression.
Collapse
Affiliation(s)
- Suleiman W Bahouth
- Department of Pharmacology, College of Medicine, the University of Tennessee Health Sciences Center, Memphis, Tennessee 38163, USA.
| | | | | |
Collapse
|
38
|
Guillemot L, Levy A, Raymondjean M, Rothhut B. Angiotensin II-induced transcriptional activation of the cyclin D1 gene is mediated by Egr-1 in CHO-AT(1A) cells. J Biol Chem 2001; 276:39394-403. [PMID: 11502738 DOI: 10.1074/jbc.m103862200] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cyclin D1 protein expression is regulated by mitogenic stimuli and is a critical component in the regulation of G(1) to S phase progression of the cell cycle. Angiotensin II (Ang II) binds to specific G protein-coupled receptors and is mitogenic in Chinese hamster ovary cells stably expressing the rat vascular Ang II type 1A receptor (CHO-AT(1A)). We recently reported that in these cells, Ang II induced cyclin D1 promoter activation and protein expression in a phosphatidylinositol 3-kinase (PI3K)-, SHP-2-, and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK)-dependent manner (Guillemot, L., Levy, A., Zhao, Z. J., Béréziat, G., and Rothhut, B. (2000) J. Biol. Chem. 275, 26349-26358). In this report, transfection studies using a series of deleted cyclin D1 promoters revealed that two regions between base pairs (bp) -136 and -96 and between bp -29 and +139 of the human cyclin D1 promoter contained regulatory elements required for Ang II-mediated induction. Mutational analysis in the -136 to -96 bp region provided evidence that a Sp1/early growth response protein (Egr) motif was responsible for cyclin D1 promoter activation by Ang II. Gel shift and supershift studies showed that Ang II-induced Egr-1 binding involved de novo protein synthesis and correlated well with Egr-1 promoter activation. Both U0126 (an inhibitor of the MAPK/ERK kinase MEK) and wortmannin (an inhibitor of PI3K) abrogated Egr-1 endogenous expression and Egr-1 promoter activity induced by Ang II. Moreover, using a co-transfection approach, we found that Ang II induction of Egr-1 promoter activity was blocked by dominant-negative p21(ras), Raf-1, and tyrosine phosphatase SHP-2 mutants. Identical effects were obtained when inhibitors and dominant negative mutants were tested on the -29 to +139 bp region of the cyclin D1 promoter. Taken together, these findings demonstrate that Ang II-induced cyclin D1 up-regulation is mediated by the activation and specific interaction of Egr-1 with the -136 to -96 bp region of the cyclin D1 promoter and by activation of the -29 to +139 bp region, both in a p21(ras)/Raf-1/MEK/ERK-dependent manner, and also involves PI3K and SHP-2.
Collapse
Affiliation(s)
- L Guillemot
- UMR Physiologie et Physiopathologie, Université Pierre et Marie Curie, Case Courrier 256, Bâtiment A, 5ème étage, 7 Quai St-Bernard, Paris 75005, France
| | | | | | | |
Collapse
|
39
|
Gousseva N, Kugathasan K, Chesterman CN, Khachigian LM. Early growth response factor-1 mediates insulin-inducible vascular endothelial cell proliferation and regrowth after injury. J Cell Biochem 2001; 81:523-34. [PMID: 11255235 DOI: 10.1002/1097-4644(20010601)81:3<523::aid-jcb1066>3.0.co;2-e] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hyperinsulinemia in diabetes mellitus is a significant risk factor in the development of atherosclerosis and early restenosis after balloon angioplasty. These manifestations could be mediated by the ability of insulin to potentiate the cellular proliferative and reparative response of vascular cell types to local stimuli. Here we demonstrate that insulin stimulates DNA synthesis in aortic endothelial cells. Reverse transcription-polymerase chain reaction and Northern blotting revealed that insulin induces the expression and transcriptional activity of the immediate early gene and zinc finger transcription protein, early growth response factor-1 (Egr-1). Western immunoblot analysis revealed that insulin-inducible Egr-1 expression was inhibited using phosphorothioate-specific antisense oligonucleotides targeting Egr-1 mRNA. These agents blocked endothelial cell DNA synthesis stimulated by insulin in a dose-dependent manner and inhibited the capacity of insulin to potentiate the reparative response of endothelial cells to mechanical injury in vitro. These oligonucleotides also attenuated wound repair in smooth muscle cells. DNA synthesis induced by insulin was suppressed by inhibitors of two upstream activators of Egr-1, extracellular signal-regulated kinase (ERK) and phosphatidylinositol 3-phosphate (PI 3-K), whereas p38 kinase inhibitors had no effect. These present findings demonstrate that insulin-inducible DNA synthesis and repair after injury are processes critically dependent upon the activation of Egr-1. Additionally, they implicate this transcription factor as a potential target for the inhibition of restenosis in diabetics.
Collapse
Affiliation(s)
- N Gousseva
- Centre for Thrombosis and Vascular Research, The University of New South Wales, Sydney NSW, Australia
| | | | | | | |
Collapse
|
40
|
Abstract
Angiotensin (Ang) II is considered a regulatory hormone stimulating vascular smooth muscle cell constriction, aldosterone release from the adrenal gland, and sodium reabsorption in the renal tubule. Furthermore, Ang II may be formed and act locally as a chemokine, inducing tyrosine phosphorylation, cell growth, hypertrophy, and differentiation. In addition, evidence has recently accrued showing that Ang II is important in stimulating the production of reactive oxygen species and the activation of ancient inflammatory mechanisms. The transcription factor nuclear factor kappa-B is pivotal to these processes. Nuclear factor kappa-B activation stimulates the expression of a gene menagerie important to chemoattraction, surface adhesion molecule expression, coagulation, and inflammation. Anti-inflammatory interventions may have therapeutic utility.
Collapse
Affiliation(s)
- F C Luft
- Franz Volhard Clinic and Max Delbrück Center for Molecular Medicine, Medical Faculty of the Charité, Humboldt University of Berlin, Germany.
| |
Collapse
|
41
|
Abstract
We are used to thinking of angiotensin (Ang) II as a regulatory hormone that stimulates constriction of vascular smooth muscle cells, aldosterone release from the adrenal gland, and sodium reabsorption in the renal tubule. We have also become accustomed to understanding that Ang II may be formed and may act locally as a chemokine that induces tyrosine phosphorylation, cell growth, hypertrophy, and differentiation. Viewing Ang II as an inflammatory molecule is stranger still. Nevertheless, recent evidence shows that Ang II is important in stimulating the production of reactive oxygen species and the activation of ancient inflammatory mechanisms. The nuclear factor kappaB (NF-kappaB) is pivotal to these processes. Activation of NF-kappaB stimulates the expression of a gene menagerie that is important to chemoattraction, expression of surface adhesion molecules, coagulation, and inflammation. In addition, Ang II has been shown to regulate cellular immune responses. It stimulates the proliferation of lymphocytes and contributes to their activation via calcineurin-related pathways. Knowledge of these mechanisms may provide additional therapeutic avenues.
Collapse
Affiliation(s)
- F C Luft
- Franz Volhard Clinic and Max Delbrück Center for Molecular Medicine, Wiltberg Strasse 50, 13125 Berlin, Germany.
| |
Collapse
|
42
|
Du B, Fu C, Kent KC, Bush H, Schulick AH, Kreiger K, Collins T, McCaffrey TA. Elevated Egr-1 in human atherosclerotic cells transcriptionally represses the transforming growth factor-beta type II receptor. J Biol Chem 2000; 275:39039-47. [PMID: 10982796 DOI: 10.1074/jbc.m005159200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Atherosclerotic lesions may progress due to a "failure to die" by vascular repair cells. Egr-1, a zinc finger transcription factor, is elevated more than 5-fold in human carotid lesions relative to the adjacent tunica media. Lesion cells in vitro also express 2-3-fold higher Egr-1 mRNA and protein levels but express much lower levels of the transforming growth factor-beta (TGF-beta) Type II receptor (TbetaR-2) and are functionally resistant to the antiproliferative effects of TGF-beta. Lesion cells fail to express a TbetaR-2 promoter/chloramphenicol acetyltransferase (CAT) construct but overexpress an Egr-1-inducible platelet-derived growth factor-A promoter/CAT construct. Transfection of Egr-1 cDNA represses TbetaR-2/CAT constructs but induces PDGF-A/CAT. Egr-1 transfection reduces the levels of TbetaR-2 and confers resistance to the antiproliferative effect of TGF-beta1. Egr-1 can interact directly with both the -143 Sp1 site and the positive regulatory element 2 (PRE2) (ERT/ets) region of the TbetaR-2 promoter. Thus, although activating a family of stress-responsive genes, Egr-1 also transcriptionally represses one of the major inhibitory pathways that restrains vascular repair.
Collapse
MESH Headings
- Arteries/metabolism
- Arteriosclerosis/metabolism
- Binding Sites
- Blotting, Western
- Cell Division
- Cell Nucleus/metabolism
- Cells, Cultured
- Chloramphenicol O-Acetyltransferase/metabolism
- Cloning, Molecular
- DNA, Complementary/metabolism
- DNA-Binding Proteins/biosynthesis
- Densitometry
- Dose-Response Relationship, Drug
- Early Growth Response Protein 1
- Fibroblast Growth Factor 2/metabolism
- Genes, Reporter
- Humans
- Immediate-Early Proteins
- Nerve Growth Factor/metabolism
- Platelet-Derived Growth Factor/metabolism
- Promoter Regions, Genetic
- Protein Serine-Threonine Kinases
- RNA/metabolism
- RNA, Messenger/metabolism
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/metabolism
- Recombinant Proteins/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Time Factors
- Transcription Factors/biosynthesis
- Transcription, Genetic
- Transfection
- Veins/metabolism
- Zinc Fingers
Collapse
Affiliation(s)
- B Du
- Department of Medicine, Division of Hematology/Oncology, Weill Medical College of Cornell University New York, New York 10021, USA
| | | | | | | | | | | | | | | |
Collapse
|