1
|
Lu Y, Yu X, Chen Y, Wu C, Jiang Q, Ha S, Zhu D, Bi Y, Liu X, Zhang H, Li Z, Wang W, Li L, Chen H, Zhang Y, Dai H, Fang J. Safety and Efficacy of Multiple Escalating Doses of RC28-E for Neovascular Age-Related Macular Degeneration: A Phase 1b Trial. Ophthalmol Ther 2024; 13:2405-2415. [PMID: 39031277 PMCID: PMC11341794 DOI: 10.1007/s40123-024-00994-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 06/27/2024] [Indexed: 07/22/2024] Open
Abstract
INTRODUCTION To assess the safety and efficacy of repeated intravitreal injections of RC28-E, a novel bispecific antibody that simultaneously binds vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in patients with neovascular age-related macular degeneration (AMD). This was a prospective, multicenter, open-label clinical trial; 37 patients with choroidal neovascularization secondary to AMD and best-corrected visual acuity (BCVA) letter scores between 73 and 34 were enrolled. METHODS Treatment regimens consisted of a 3-month loading phase and a pro re nata (PRN) maintenance phase. This study included three treatment groups: the 0.5, 1.0, and 2.0 mg RC28-E groups, with escalating doses ranging from 0.5 to 2.0 mg. Patients were evaluated monthly for 48 weeks. Safety was assessed based on ocular and systemic adverse events (AEs), pharmacokinetic characteristics, and the presence of anti-RC28-E antibodies. Efficacy was assessed using the mean change in BCVA and central subfield thickness (CST) from baseline to week 48. RESULTS Most AEs were mild or moderate. The most common AE was a minor injection-related subconjunctival hemorrhage (16.2%). The AEs did not increase with dose or repeated injections. At week 48, mean improvements in BCVA from baseline in the 0.5, 1.0, and 2.0 mg groups were 6.1 ± 8.3, 9.9 ± 10.7, and 7.6 ± 9.38 letters, respectively; mean reductions in CST in the three groups were 112.1 ± 160.5, 175.1 ± 212.4, and 128.7 ± 145.8 μm, respectively. The serum RC28-E concentrations in 95% of the patients were below the quantification limit of the assay. No significant change from baseline was observed in the mean plasma concentrations of VEGF or FGF over the 48 weeks of treatment. Pre-treatment antibodies to RC28-E were detected in 1 of the 37 patients. Antibodies to RC28-E were detected in two patients after dosing with RC28-E for 48 weeks. CONCLUSION RC28-E was well tolerated and exhibited an overall favorable safety profile with evidence of improvements in BCVA and anatomical parameters.
Collapse
Affiliation(s)
- Yingyi Lu
- Department of Ophthalmology, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaobing Yu
- Department of Ophthalmology, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Youxin Chen
- Department of Ophthalmology, Union Medical College Hospital, Chinese Academy of Medical Sciences, PekingBeijing, China
| | - Chan Wu
- Department of Ophthalmology, Union Medical College Hospital, Chinese Academy of Medical Sciences, PekingBeijing, China
| | - Qin Jiang
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shaoping Ha
- Department of Ophthalmology, The People's Hospital of Ningxia Autonomous Region, Yinchuan, Ningxia, China
| | - Dan Zhu
- Department of Ophthalmology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yanlong Bi
- Department of Ophthalmology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xiaoling Liu
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Han Zhang
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhuo Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | | | - Lin Li
- RemeGen Co., Ltd, Yantai, Shandong, China
| | - He Chen
- RemeGen Co., Ltd, Yantai, Shandong, China
| | | | - Hong Dai
- Department of Ophthalmology, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
2
|
Yang W, Zhou X, Li Q, Yin M, Wang N. The effect of overexpression of CyPA on gene expression in human umbilical vein endothelial cells. Medicine (Baltimore) 2024; 103:e38886. [PMID: 39029007 PMCID: PMC11398797 DOI: 10.1097/md.0000000000038886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2024] Open
Abstract
The aim of this study is to screen the differentially expressed genes and genes with alternative splicing in PPIA overexpressing cells by transcriptome sequencing. Transcriptome sequencing was performed to identify differentially expressed genes and genes with altered alternative splicing in PPIA overexpressing cells and results were validated by real-time quantitative polymerase chain reaction. The biological function and pathways of those genes were further explored through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes analyses. A total of 157 significantly upregulated genes and 171 significantly downregulated genes were identified in PPIA overexpressing cells, and the splicing pattern of LHPP, APH1A, BRD1, and ORAI3 was found to be altered. GO analyses showed that the most enriched GO terms of the 157 upregulated genes included extracellular region, protein binding, and metal ion, and the most enriched GO terms of the 171 downregulated genes included binding neuron projection, protein binding, and endoplasmic reticulum unfolded protein response. Kyoto Encyclopedia of Genes and Genomes analyses showed that the 157 upregulated genes were mainly enriched in gastric acid secretion, Mitogen-activated protein kinase signaling pathway, etc, and the 171 downregulated genes were mainly enriched in transcriptional misregulation in cancer, Tumor necrosis factor signaling pathway, etc. The overexpression of PPIA in human umbilical vein endothelial cells causes changes in the expression of downstream genes and induces alternative splicing in multiple genes. PPIA alters the expression or the alternative splicing pattern of downstream genes, leading to pathogenesis of vascular endothelial injury by high glucose mediated through CyPA.
Collapse
Affiliation(s)
- Wenwen Yang
- The First Department of General Internal Medicine, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - XinRong Zhou
- The Coronary Heart Disease Care Unit, CCU, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qiuju Li
- The First Department of General Internal Medicine, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Mingyue Yin
- The First Department of General Internal Medicine, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ning Wang
- The First Department of General Internal Medicine, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia
| |
Collapse
|
3
|
Pereira DS, Akita K, Bhisitkul RB, Nishihata T, Ali Y, Nakamura E, Nakamura Y. Safety and tolerability of intravitreal umedaptanib pegol (anti-FGF2) for neovascular age-related macular degeneration (nAMD): a phase 1, open-label study. Eye (Lond) 2024; 38:1149-1154. [PMID: 38040965 PMCID: PMC11009303 DOI: 10.1038/s41433-023-02849-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 12/03/2023] Open
Abstract
OBJECTIVE To evaluate the efficacy and safety of a single-dose intravitreal umedaptanib pegol (anti-FGF2, investigational new drug) for the treatment of neovascular age-related macular degeneration (nAMD). METHODS Nine participants who had a diagnosis of refractory nAMD were enrolled and received a single intravitreal injection of umedaptanib pegol at increasing doses of 0.2, 1.0 or 2.0 mg in the study eye. RESULTS All three doses of umedaptanib pegol evaluated in the study were safe and well tolerated. No severe adverse event (AE) was observed in the study. There was an improvement in retinal fluid measured by central subfield thickness (CST) in most subjects. Remarkably, all three subjects who received 2.0 mg/eye showed improvement of more than 150 μm. CONCLUSIONS Intravitreal umedaptanib pegol was safe, well tolerated, and demonstrated an indication of bioactivity in participants that have persistent subretinal fluid refractory to the treatment with anti-VEGFs.
Collapse
Affiliation(s)
| | | | - Robert B Bhisitkul
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | - Yoshikazu Nakamura
- RIBOMIC Inc., Minato-ku, Tokyo, Japan.
- Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan.
| |
Collapse
|
4
|
Nakamura Y. Multiple Therapeutic Applications of RBM-007, an Anti-FGF2 Aptamer. Cells 2021; 10:cells10071617. [PMID: 34203430 PMCID: PMC8305614 DOI: 10.3390/cells10071617] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) plays a pivotal role in angiogenesis, but is not the only player with an angiogenic function. Fibroblast growth factor-2 (FGF2), which was discovered before VEGF, is also an angiogenic growth factor. It has been shown that FGF2 plays positive pathophysiological roles in tissue remodeling, bone health, and regeneration, such as the repair of neuronal damage, skin wound healing, joint protection, and the control of hypertension. Targeting FGF2 as a therapeutic tool in disease treatment through clinically useful inhibitors has not been developed until recently. An isolated inhibitory RNA aptamer against FGF2, named RBM-007, has followed an extensive preclinical study, with two clinical trials in phase 2 and phase 1, respectively, underway to assess the therapeutic impact in age-related macular degeneration (wet AMD) and achondroplasia (ACH), respectively. Moreover, showing broad therapeutic potential, preclinical evidence supports the use of RBM-007 in the treatment of lung cancer and cancer pain.
Collapse
Affiliation(s)
- Yoshikazu Nakamura
- Division of RNA Medical Science, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan;
- RIBOMIC Inc., Tokyo 108-0071, Japan
| |
Collapse
|
5
|
Nakada EM, Sun R, Fujii U, Martin JG. The Impact of Endoplasmic Reticulum-Associated Protein Modifications, Folding and Degradation on Lung Structure and Function. Front Physiol 2021; 12:665622. [PMID: 34122136 PMCID: PMC8188853 DOI: 10.3389/fphys.2021.665622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022] Open
Abstract
The accumulation of unfolded/misfolded proteins in the endoplasmic reticulum (ER) causes ER stress and induces the unfolded protein response (UPR) and other mechanisms to restore ER homeostasis, including translational shutdown, increased targeting of mRNAs for degradation by the IRE1-dependent decay pathway, selective translation of proteins that contribute to the protein folding capacity of the ER, and activation of the ER-associated degradation machinery. When ER stress is excessive or prolonged and these mechanisms fail to restore proteostasis, the UPR triggers the cell to undergo apoptosis. This review also examines the overlooked role of post-translational modifications and their roles in protein processing and effects on ER stress and the UPR. Finally, these effects are examined in the context of lung structure, function, and disease.
Collapse
Affiliation(s)
- Emily M. Nakada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - Rui Sun
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - Utako Fujii
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - James G. Martin
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| |
Collapse
|
6
|
Geng W, Ren J, Shi H, Qin F, Xu X, Xiao S, Jiao Y, Wang A. RPL41 sensitizes retinoblastoma cells to chemotherapeutic drugs via ATF4 degradation. J Cell Physiol 2020; 236:2214-2225. [PMID: 32783256 DOI: 10.1002/jcp.30010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 07/07/2020] [Accepted: 08/01/2020] [Indexed: 01/03/2023]
Abstract
Retinoblastoma is the most common intraocular cancer with metastatic potential affecting infants and children. Although chemotherapy is available for retinoblastoma, side effects and drug resistance are frequent. Rpl41, encoding ribosomal protein L41 (RPL41), has been identified as a tumor suppressor gene, and its targeted degradation of activating transcription factor 4 (ATF4) produces an antitumor effect. The goal of the present study is to provide experimental evidence for the clinical application of a small peptide regimen in combination with chemotherapy for the treatment of retinoblastoma and to investigate the mechanism of their combined cytotoxicity. It was observed that treatment with the RPL41 peptide alone decreased the viability, migration, and invasion of retinoblastoma Y79 and Weri-Rb1 cells, in addition to promoting cell apoptosis and cell cycle arrest. Furthermore, RPL41 protein levels showed a significantly decreased trend in retinoblastoma specimens, whereas ATF4 protein levels tended to be increased. Mechanistically, ATF4 degradation as a result of RPL41 peptide treatment was observed in retinoblastoma Y79 and Weri-Rb1 cells. Most important, low-dose administration of the RPL41 peptide significantly enhanced the antitumor effect of carboplatin, and further analysis confirmed their synergistic effect as anti-retinoblastoma therapy, indicating that RPL41 sensitized Y79 and Weri-Rb1 retinoblastoma cells to carboplatin. Thus, our data provide a preclinical rationale for the exploration of the RPL41 peptide as a potential adjuvant to carboplatin treatment in retinoblastoma.
Collapse
Affiliation(s)
- Wen Geng
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shengyang, Liaoning, China
| | - Jiaxu Ren
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shengyang, Liaoning, China
| | - Huimin Shi
- Department of Ophthalmology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Feng Qin
- Department of Ophthamology, Shenyang Aier Eye Hospital, Shenyang, Liaoning, China
| | - Xiaohe Xu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shengyang, Liaoning, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women's Hospital of Harvard Medical School, Boston, Massachusetts
| | - Yisheng Jiao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shengyang, Liaoning, China
| | - Aiyuan Wang
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shengyang, Liaoning, China
| |
Collapse
|
7
|
Starvation and antimetabolic therapy promote cytokine release and recruitment of immune cells. Proc Natl Acad Sci U S A 2020; 117:9932-9941. [PMID: 32312819 PMCID: PMC7211964 DOI: 10.1073/pnas.1913707117] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Cellular starvation is typically a consequence of tissue injury that disrupts the local blood supply but can also occur where cell populations outgrow the local vasculature, as observed in solid tumors. Cells react to nutrient deprivation by adapting their metabolism, or, if starvation is prolonged, it can result in cell death. Cell starvation also triggers adaptive responses, like angiogenesis, that promote tissue reorganization and repair, but other adaptive responses and their mediators are still poorly characterized. To explore this issue, we analyzed secretomes from glucose-deprived cells, which revealed up-regulation of multiple cytokines and chemokines, including IL-6 and IL-8, in response to starvation stress. Starvation-induced cytokines were cell type-dependent, and they were also released from primary epithelial cells. Most cytokines were up-regulated in a manner dependent on NF-κB and the transcription factor of the integrated stress response ATF4, which bound directly to the IL-8 promoter. Furthermore, glutamine deprivation, as well as the antimetabolic drugs 2-deoxyglucose and metformin, also promoted the release of IL-6 and IL-8. Finally, some of the factors released from starved cells induced chemotaxis of B cells, macrophages, and neutrophils, suggesting that nutrient deprivation in the tumor environment can serve as an initiator of tumor inflammation.
Collapse
|
8
|
Riascos-Bernal DF. Perking Up Strategies to Control Restenosis. JACC Basic Transl Sci 2020; 5:264-266. [PMID: 32215377 PMCID: PMC7091502 DOI: 10.1016/j.jacbts.2020.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Dario F. Riascos-Bernal
- Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
9
|
PERK Inhibition Mitigates Restenosis and Thrombosis: A Potential Low-Thrombogenic Antirestenotic Paradigm. JACC Basic Transl Sci 2020; 5:245-263. [PMID: 32215348 PMCID: PMC7091514 DOI: 10.1016/j.jacbts.2019.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 12/13/2019] [Accepted: 12/13/2019] [Indexed: 12/18/2022]
Abstract
Drug-eluting stents impede neointimal smooth muscle cell hyperplasia but exacerbate endothelial cell dysfunction and thrombogenicity. It has been a challenge to identify a common target to inhibit both. Findings in this study suggest PERK as such a target. A PERK inhibitor administered either via an endovascular (in biomimetic nanocarriers) or perivascular (in hydrogel) route effectively mitigated neointimal hyperplasia in rats. Oral gavage of the PERK inhibitor partially preserved the normal blood flow in a mouse model of induced thrombosis. Dampening PERK activity inhibited STAT3 while activating SRF in smooth muscle cells, and also reduced prothrombogenic tissue factor and growth impairment of endothelial cells.
Developing endothelial-protective, nonthrombogenic antirestenotic treatments has been a challenge. A major hurdle to this has been the identification of a common molecular target in both smooth muscle cells and endothelial cells, inhibition of which blocks dysfunction of both cell types. The authors’ findings suggest that the PERK kinase could be such a target. Importantly, PERK inhibition mitigated both restenosis and thrombosis in preclinical models, implicating a low-thrombogenic antirestenotic paradigm.
Collapse
Key Words
- ATF, activating transcription factor
- Ad, adenovirus
- CHOP, CCAAT-enhancer-binding protein homologous protein
- DES, drug-eluting stents
- DMSO, dimethyl sulfoxide
- EC, endothelial cell
- ER, endoplasmic reticulum
- FBS, fetal bovine serum
- GFP, green fluorescent protein
- HA, hemagglutinin
- I/M, intima to media
- IEL, internal elastic lamina
- IH, intimal hyperplasia
- IRE1, inositol-requiring kinase 1
- MRTF-A, myocardin related transcription factor A
- PDGF, platelet-derived growth factor
- PDGF-BB, platelet-derived growth factor with 2 B subunits
- PERK
- PERK, protein kinase RNA-like endoplasmic reticulum kinase
- SMA, smooth muscle actin
- SMC, smooth muscle cell
- SRF, serum response factor
- STAT3, signal transducer and activator of transcription 3
- TNF, tumor necrosis factor
- eIF2, eukaryotic translation initiation factor 2
- endothelial cells
- restenosis
- siRNA, small interfering ribonucleic acid
- smooth muscle cells
- thrombosis
Collapse
|
10
|
Xue CD, Chen Y, Ren JL, Zhang LS, Liu X, Yu YR, Tang CS, Qi YF. Endogenous intermedin protects against intimal hyperplasia by inhibiting endoplasmic reticulum stress. Peptides 2019; 121:170131. [PMID: 31408662 DOI: 10.1016/j.peptides.2019.170131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/27/2019] [Accepted: 08/05/2019] [Indexed: 12/15/2022]
Abstract
Extensive proliferation of vascular smooth muscle cell (VSMC) contributes to intimal hyperplasia following vascular injury, in which endoplasmic reticulum stress (ERS) plays a critical role. Intermedin (IMD) is a vascular paracrine/autocrine peptide exerting numerous beneficial effects in cardiovascular diseases. IMD overexpression could alleviate intimal hyperplasia. Here, we investigated whether endogenous IMD protects against intimal hyperplasia by inhibiting endoplasmic reticulum stress. The mouse left common carotid-artery ligation-injury model was established to induce intimal hyperplasia using IMD-/-mice and C57BL/6 J wild-type (WT) mice. Platelet-derived growth factor-BB (PDGF-BB) was used to stimulate the proliferation of VSMC. IMD-/- mice displayed exacerbated intimal hyperplasia induced by complete ligation of the left carotid artery at 14 d and 28 d compared to WT mice. However, IMD-deficiency had no effect on blood pressure, plasma triglyceride, and fasting blood glucose levels in mice. Furthermore, VSMCs derived from IMD-/- mice showed increased cell proliferation and dramatically elevated levels of glucose regulated protein 78 (GRP78), activating transcription factor 4 (ATF4), ATF6 mRNA under PDGF-BB treatment compared to WT mice-derived VSMCs. In addition, exogenous administration of IMD significantly attenuated PDGF-BB-induced cell proliferation and GRP78, phosphorylase-inositol requiring enzyme 1α, ATF4, and ATF6 protein levels. Thus, endogenous IMD may counteract ERS to exert protective role in response to vascular injury and IMD is expected to be a therapeutic target for the prevention and treatment of restenosis.
Collapse
MESH Headings
- Activating Transcription Factor 4
- Activating Transcription Factor 6/genetics
- Activating Transcription Factor 6/metabolism
- Animals
- Becaplermin/pharmacology
- Carotid Arteries/surgery
- Cell Proliferation/drug effects
- Disease Models, Animal
- Endoplasmic Reticulum Chaperone BiP
- Endoplasmic Reticulum Stress/drug effects
- Endoplasmic Reticulum Stress/genetics
- Gene Expression Regulation
- Heat-Shock Proteins
- Hyperplasia/genetics
- Hyperplasia/metabolism
- Hyperplasia/pathology
- Hyperplasia/prevention & control
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Neuropeptides/deficiency
- Neuropeptides/genetics
- Primary Cell Culture
- Signal Transduction
- Tunica Intima/metabolism
- Tunica Intima/pathology
Collapse
Affiliation(s)
- Chang-Ding Xue
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Yao Chen
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Jin-Ling Ren
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Lin-Shuang Zhang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Xin Liu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Yan-Rong Yu
- Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Chao-Shu Tang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China
| | - Yong-Fen Qi
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China.
| |
Collapse
|
11
|
Matsuda Y, Nonaka Y, Futakawa S, Imai H, Akita K, Nishihata T, Fujiwara M, Ali Y, Bhisitkul RB, Nakamura Y. Anti-Angiogenic and Anti-Scarring Dual Action of an Anti-Fibroblast Growth Factor 2 Aptamer in Animal Models of Retinal Disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:819-828. [PMID: 31454678 PMCID: PMC6716068 DOI: 10.1016/j.omtn.2019.07.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 06/25/2019] [Accepted: 07/24/2019] [Indexed: 12/03/2022]
Abstract
Currently approved therapies for age-related macular degeneration (AMD) are inhibitors against vascular endothelial growth factor (VEGF), which is a major contributor to the pathogenesis of neovascular AMD (nAMD). Intravitreal injections of anti-VEGF drugs have shown dramatic visual benefits for AMD patients. However, a significant portion of AMD patients exhibit an incomplete response to therapy and, over the extended management course, can lose vision, with the formation of submacular fibrosis as one risk factor. We investigated a novel target for AMD treatments, fibroblast growth factor 2 (FGF2), which has been implicated in the pathophysiology of both angiogenesis and fibrosis in a variety of tissue and organ systems. The anti-FGF2 aptamer, RBM-007, was examined for treatment of nAMD in animal models. In in vivo studies conducted in mice and rats, RBM-007 was able to inhibit FGF2-induced angiogenesis, laser-induced choroidal neovascularization (CNV), and CNV with fibrosis. Pharmacokinetic studies of RBM-007 in the rabbit vitreous revealed high and relatively long-lasting profiles that are superior to other approved anti-VEGF drugs. The anti-angiogenic and anti-scarring dual action of RBM-007 holds promise as an additive or alternative therapy to anti-VEGF treatments for nAMD.
Collapse
Affiliation(s)
- Yusaku Matsuda
- RIBOMIC, Inc., 3-16-13 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan
| | - Yosuke Nonaka
- RIBOMIC, Inc., 3-16-13 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan
| | - Satoshi Futakawa
- RIBOMIC, Inc., 3-16-13 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan
| | - Hirotaka Imai
- RIBOMIC, Inc., 3-16-13 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan
| | - Kazumasa Akita
- RIBOMIC, Inc., 3-16-13 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan
| | | | | | - Yusuf Ali
- RIBOMIC, Inc., 3-16-13 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan
| | - Robert B Bhisitkul
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Yoshikazu Nakamura
- RIBOMIC, Inc., 3-16-13 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan; Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|
12
|
Ishii T, Warabi E, Mann GE. Circadian control of BDNF-mediated Nrf2 activation in astrocytes protects dopaminergic neurons from ferroptosis. Free Radic Biol Med 2019; 133:169-178. [PMID: 30189266 DOI: 10.1016/j.freeradbiomed.2018.09.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/20/2018] [Accepted: 09/01/2018] [Indexed: 01/19/2023]
Abstract
Astrocyte-neuron interactions protect neurons from iron-mediated toxicity. As dopamine can be metabolized to reactive quinones, dopaminergic neurons are susceptible to oxidative damage and ferroptosis-like induced cell death. Detoxification enzymes are required to protect neurons. Brain-derived neurotrophic factor (BDNF) plays a key role in the regulation of redox sensitive transcription factor Nrf2 in astrocytes and metabolic cooperation between astrocytes and neurons. This article reviews the importance of BDNF and astrocyte-neuron interactions in the protection of neurons against oxidative damages in rodent brains. We previously proposed that BDNF activates Nrf2 via the truncated TrkB.T1 and p75NTR receptor complex in astrocytes. Stimulation by BDNF generates the signaling molecule ceramide, which activates PKCζ leading to induction of the CK2-Nrf2 signaling axis. As a cell clock regulates p75NTR expression, we suggested that BDNF effectively activates Nrf2 in astrocytes during the rest phase. In contrast, neurons express both TrkB.FL and TrkB.T1, and TrkB.FL tyrosine kinase activity inhibits p75NTR-dependent ceramide generation and internalizes p75NTR. Therefore, BDNF may not effectively activate Nrf2 in neurons. Notably, neurons only weakly activate detoxification and antioxidant enzymes/proteins via the Nrf2-ARE signaling axis. Thus, astrocytes may provide relevant transcripts and/or proteins to neurons via microparticles/exosomes increasing neuronal resistance to oxidative stress. Circadian increases in the levels of circulating glucocorticoids may further facilitate material transfer from astrocytes to neurons via the stimulation of pannexin 1 channels-P2X7R signaling pathway in astrocytes at the beginning of the active phase. Dysregulation of astrocyte-neuron interactions could therefore contribute to the pathogenesis of neurodegenerative diseases including Parkinson's disease.
Collapse
Affiliation(s)
- Tetsuro Ishii
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan.
| | - Eiji Warabi
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
| | - Giovanni E Mann
- School of Cardiovascular Medicine and Sciences, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London SE1 9NH, UK
| |
Collapse
|
13
|
Huo JF, Chen XB. P2X4R silence suppresses glioma cell growth through BDNF/TrkB/ATF4 signaling pathway. J Cell Biochem 2018; 120:6322-6329. [PMID: 30362154 DOI: 10.1002/jcb.27919] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/27/2018] [Indexed: 02/06/2023]
Abstract
Purinergic receptor P2X 4 (P2X4R), a member of purinergic channels family and a subtype of ionotropic adenosine triphosphate receptors, plays a critical role in tumorigenesis. Evidence suggested that P2X4R is expressed in rat C6 glioma model, however, its role and the underlying mechanism of action are still unclear in human glioblastoma multiforme (GBM). In the current study, our aim is to examine the function and the molecular basis of P2X4R in GBM. We first observed that GBM cells, U251, T98, U87, U373, and A172 were all high expressed P2X4R, when compared with the normal human astrocytes (NHA) cells. To gain the function of P2X4R, P2X4R silence cells were constructed by transfection with P2X4R small interfering RNA (siRNA). We found that P2X4R deletion impeded T98 and U87 cell viability and proliferation, and further studies indicated that cell apoptosis and caspase-3 activity was increased in T98 and U87 cell transfected with P2X4R siRNA. Subsequently, we confirmed that P2X4R silence suppressed brain-derived neurotrophic factor (BDNF), Trk receptor tyrosine kinases (TrkB), and activating transcription factor 4 (ATF4) expression in T98 and U87 cells. And P2X4R siRNA-induced ATF4-expression inhibition dependent on BDNF/TrkB signaling pathway. The impact of P2X4R silence on T98 and U87 cell growth and apoptosis was reversed by ATF4 overexpression. In summary, this study provides the first evidence that P2X4R plays important roles in GBM cell growth and apoptosis.
Collapse
Affiliation(s)
- Jun-Feng Huo
- Second Ward, Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Xiao-Bing Chen
- Second Ward, Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
14
|
Alhendi AMN, Patrikakis M, Daub CO, Kawaji H, Itoh M, de Hoon M, Carninci P, Hayashizaki Y, Arner E, Khachigian LM. Promoter Usage and Dynamics in Vascular Smooth Muscle Cells Exposed to Fibroblast Growth Factor-2 or Interleukin-1β. Sci Rep 2018; 8:13164. [PMID: 30177712 PMCID: PMC6120868 DOI: 10.1038/s41598-018-30702-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/03/2018] [Indexed: 01/22/2023] Open
Abstract
Smooth muscle cells (SMC) in blood vessels are normally growth quiescent and transcriptionally inactive. Our objective was to understand promoter usage and dynamics in SMC acutely exposed to a prototypic growth factor or pro-inflammatory cytokine. Using cap analysis gene expression (FANTOM5 project) we report differences in promoter dynamics for immediate-early genes (IEG) and other genes when SMC are exposed to fibroblast growth factor-2 or interleukin-1β. Of the 1871 promoters responding to FGF2 or IL-1β considerably more responded to FGF2 (68.4%) than IL-1β (18.5%) and 13.2% responded to both. Expression clustering reveals sets of genes induced, repressed or unchanged. Among IEG responding rapidly to FGF2 or IL-1β were FOS, FOSB and EGR-1, which mediates human SMC migration. Motif activity response analysis (MARA) indicates most transcription factor binding motifs in response to FGF2 were associated with a sharp induction at 1 h, whereas in response to IL-1β, most motifs were associated with a biphasic change peaking generally later. MARA revealed motifs for FOS_FOS{B,L1}_JUN{B,D} and EGR-1..3 in the cluster peaking 1 h after FGF2 exposure whereas these motifs were in clusters peaking 1 h or later in response to IL-1β. Our findings interrogating CAGE data demonstrate important differences in promoter usage and dynamics in SMC exposed to FGF2 or IL-1β.
Collapse
Affiliation(s)
- Ahmad M N Alhendi
- Vascular Biology and Translational Research, School of Medical Sciences, University of New South Wales, Sydney, 2052, Australia
| | - Margaret Patrikakis
- Vascular Biology and Translational Research, School of Medical Sciences, University of New South Wales, Sydney, 2052, Australia
| | - Carsten O Daub
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies) (CLST DGT), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- Department of Biosciences and Nutrition and Science for Life Laboratory, Karolinska Institutet, SE-141 86, Stockholm, Sweden
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
| | - Hideya Kawaji
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies) (CLST DGT), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- RIKEN Omics Science Center (OSC), 1-7-22 Suehiro-cho, Tsurumi-ku Yokohama, 230-0045, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program (PMI), 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
- Preventive Medicine and Applied Genomics Unit, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
| | - Masayoshi Itoh
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies) (CLST DGT), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- RIKEN Omics Science Center (OSC), 1-7-22 Suehiro-cho, Tsurumi-ku Yokohama, 230-0045, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program (PMI), 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Michiel de Hoon
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies) (CLST DGT), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- RIKEN Omics Science Center (OSC), 1-7-22 Suehiro-cho, Tsurumi-ku Yokohama, 230-0045, Japan
- Laboratory for Applied Computational Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
| | - Piero Carninci
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies) (CLST DGT), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- RIKEN Omics Science Center (OSC), 1-7-22 Suehiro-cho, Tsurumi-ku Yokohama, 230-0045, Japan
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
| | - Yoshihide Hayashizaki
- RIKEN Omics Science Center (OSC), 1-7-22 Suehiro-cho, Tsurumi-ku Yokohama, 230-0045, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program (PMI), 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Erik Arner
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies) (CLST DGT), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- RIKEN Omics Science Center (OSC), 1-7-22 Suehiro-cho, Tsurumi-ku Yokohama, 230-0045, Japan
- Laboratory for Applied Regulatory Genomics Network Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
| | - Levon M Khachigian
- Vascular Biology and Translational Research, School of Medical Sciences, University of New South Wales, Sydney, 2052, Australia.
| |
Collapse
|
15
|
Liu J, Amar F, Corona C, So RWL, Andrews SJ, Nagy PL, Shelanski ML, Greene LA. Brain-Derived Neurotrophic Factor Elevates Activating Transcription Factor 4 (ATF4) in Neurons and Promotes ATF4-Dependent Induction of Sesn2. Front Mol Neurosci 2018; 11:62. [PMID: 29599707 PMCID: PMC5863619 DOI: 10.3389/fnmol.2018.00062] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/14/2018] [Indexed: 12/17/2022] Open
Abstract
Activating transcription factor 4 (ATF4) plays important physiologic roles in the brain including regulation of learning and memory as well as neuronal survival and death. Yet, outside of translational regulation by the eIF2α-dependent stress response pathway, there is little information about how its levels are controlled in neurons. Here, we show that brain-derived neurotrophic factor (BDNF) promotes a rapid and sustained increase in neuronal ATF4 transcripts and protein levels. This increase is dependent on tropomyosin receptor kinase (TrkB) signaling, but independent of levels of phosphorylated eIF2α. The elevation in ATF4 protein occurs both in nuclei and processes. Transcriptome analysis revealed that ATF4 mediates BDNF-promoted induction of Sesn2 which encodes Sestrin2, a protector against oxidative and genotoxic stresses and a mTor complex 1 inhibitor. In contrast, BDNF-elevated ATF4 did not affect expression of a number of other known ATF4 targets including several with pro-apoptotic activity. The capacity of BDNF to elevate neuronal ATF4 may thus represent a means to maintain this transcription factor at levels that provide neuroprotection and optimal brain function without risk of triggering neurodegeneration.
Collapse
Affiliation(s)
- Jin Liu
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Fatou Amar
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Carlo Corona
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Raphaella W L So
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Stuart J Andrews
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Peter L Nagy
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Michael L Shelanski
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Lloyd A Greene
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, United States
| |
Collapse
|
16
|
Cai W, Salvador-Reyes LA, Zhang W, Chen QY, Matthew S, Ratnayake R, Seo SJ, Dolles S, Gibson DJ, Paul VJ, Luesch H. Apratyramide, a Marine-Derived Peptidic Stimulator of VEGF-A and Other Growth Factors with Potential Application in Wound Healing. ACS Chem Biol 2018; 13:91-99. [PMID: 29205032 DOI: 10.1021/acschembio.7b00827] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A novel linear depsipeptide enriched with tyrosine-derived moieties, termed apratyramide, was isolated from an apratoxin-producing cyanobacterium. The structure was determined using a combination of NMR spectroscopy, mass spectrometry, and chiral analysis of the acid hydrolyzate and confirmed by total synthesis. Apratyramide up-regulated multiple growth factors at the transcript level in human keratinocyte (HaCaT) cells and induced the secretion of vascular endothelial growth factor A (VEGF-A) from HaCaT cells, suggesting the compound's potential wound-healing properties through growth factor induction. Transcriptome analysis and sequential validation supported the hypothesis and indicated its mode of action (MOA) through the unfolded protein response (UPR) pathway, which is functionally related to wound healing and angiogenesis. The conditioned medium of HaCaT cells treated with apratyramide induced angiogenesis in vitro. An ex vivo rabbit corneal epithelial model was applied to confirm the VEGF-A induction in this wound-healing model.
Collapse
Affiliation(s)
| | - Lilibeth A. Salvador-Reyes
- Marine
Science Institute, College of Science, University of the Philippines, Diliman, Quezon
City 1100, Philippines
| | - Wei Zhang
- School
of Pharmacy, Fudan University, Shanghai 200433, China
| | | | | | | | | | | | | | - Valerie J. Paul
- Smithsonian Marine Station, Fort Pierce, Florida 34949, United States
| | | |
Collapse
|
17
|
Cilostazol attenuates intimal hyperplasia in a mouse model of chronic kidney disease. PLoS One 2017; 12:e0187872. [PMID: 29206849 PMCID: PMC5716535 DOI: 10.1371/journal.pone.0187872] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 10/29/2017] [Indexed: 11/21/2022] Open
Abstract
Intimal hyperplasia (IH) is a common cause of vasculopathy due to direct endothelial damage (such as post-coronary revascularization) or indirect injury (such as chronic kidney disease, or CKD). Although the attenuation of coronary revascularization-induced IH (direct-vascular-injury-induced IH) by cilostazol, a phosphodiesterase III inhibitor, has been demonstrated, our understanding of the effect on CKD-induced IH (indirect-vascular-injury-induced IH) is limited. Herein, we tested if cilostazol attenuated CKD-induced IH in a mouse model of ischemic-reperfusion injury with unilateral nephrectomy (Chr I/R), a normotensive non-proteinuria CKD model. Cilostazol (50 mg/kg/day) or placebo was orally administered once daily from 1-week post-nephrectomy. At 20 weeks, cilostazol significantly attenuated aortic IH as demonstrated by a 34% reduction in the total intima area with 50% and 47% decreases in the ratios of tunica intima area/tunica media area and tunica intima area/(tunica intima + tunica media area), respectively. The diameters of aorta and renal function were unchanged by cilostazol. Interestingly, cilostazol decreased miR-221, but enhanced miR-143 and miR-145 in either in vitro or aortic tissue, as well as attenuated several pro-inflammatory mediators, including asymmetrical dimethylarginine, high-sensitivity C-reactive protein, vascular endothelial growth factor in aorta and serum pro-inflammatory cytokines (IL-6 and TNF-α). We demonstrated a proof of concept of the effectiveness of cilostazol in attenuating IH in a Chr I/R mouse model, a CKD model with predominantly indirect-vascular-injury-induced IH. These considerations warrant further investigation to develop a new primary prevention strategy for CKD-related IH.
Collapse
|
18
|
Nabzdyk CS, Pradhan-Nabzdyk L, LoGerfo FW. RNAi therapy to the wall of arteries and veins: anatomical, physiologic, and pharmacological considerations. J Transl Med 2017; 15:164. [PMID: 28754174 PMCID: PMC5534068 DOI: 10.1186/s12967-017-1270-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/20/2017] [Indexed: 12/02/2022] Open
Abstract
Background Cardiovascular disease remains a major health care challenge. The knowledge about the underlying mechanisms of the respective vascular disease etiologies has greatly expanded over the last decades. This includes the contribution of microRNAs, endogenous non-coding RNA molecules, known to vastly influence gene expression. In addition, short interference RNA has been established as a mechanism to temporarily affect gene expression. This review discusses challenges relating to the design of a RNA interference therapy strategy for the modulation of vascular disease. Despite advances in medical and surgical therapies, atherosclerosis (ATH), aortic aneurysms (AA) are still associated with high morbidity and mortality. In addition, intimal hyperplasia (IH) remains a leading cause of late vein and prosthetic bypass graft failure. Pathomechanisms of all three entities include activation of endothelial cells (EC) and dedifferentiation of vascular smooth muscle cells (VSMC). RNA interference represents a promising technology that may be utilized to silence genes contributing to ATH, AA or IH. Successful RNAi delivery to the vessel wall faces multiple obstacles. These include the challenge of cell specific, targeted delivery of RNAi, anatomical barriers such as basal membrane, elastic laminae in arterial walls, multiple layers of VSMC, as well as adventitial tissues. Another major decision point is the route of delivery and potential methods of transfection. A plethora of transfection reagents and adjuncts have been described with varying efficacies and side effects. Timing and duration of RNAi therapy as well as target gene choice are further relevant aspects that need to be addressed in a temporo-spatial fashion. Conclusions While multiple preclinical studies reported encouraging results of RNAi delivery to the vascular wall, it remains to be seen if a single target can be sufficient to the achieve clinically desirable changes in the injured vascular wall in humans. It might be necessary to achieve simultaneous and/or sequential silencing of multiple, synergistically acting target genes. Some advances in cell specific RNAi delivery have been made, but a reliable vascular cell specific transfection strategy is still missing. Also, off-target effects of RNAi and unwanted effects of transfection agents on gene expression are challenges to be addressed. Close collaborative efforts between clinicians, geneticists, biologists, and chemical and medical engineers will be needed to provide tailored therapeutics for the various types of vascular diseases.
Collapse
Affiliation(s)
- Christoph S Nabzdyk
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Frank W. LoGerfo Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA
| | - Leena Pradhan-Nabzdyk
- Frank W. LoGerfo Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA.
| | - Frank W LoGerfo
- Frank W. LoGerfo Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA
| |
Collapse
|
19
|
Lai DW, Lin KH, Sheu WHH, Lee MR, Chen CY, Lee WJ, Hung YW, Shen CC, Chung TJ, Liu SH, Sheu ML. TPL2 (Therapeutic Targeting Tumor Progression Locus-2)/ATF4 (Activating Transcription Factor-4)/SDF1α (Chemokine Stromal Cell-Derived Factor-α) Axis Suppresses Diabetic Retinopathy. Circ Res 2017; 121:e37-e52. [PMID: 28724746 DOI: 10.1161/circresaha.117.311066] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/09/2017] [Accepted: 07/19/2017] [Indexed: 11/16/2022]
Abstract
RATIONALE Diabetic retinopathy is characterized by vasopermeability, vascular leakage, inflammation, blood-retinal barrier breakdown, capillary degeneration, and neovascularization. However, the mechanisms underlying the association between diabetes mellitus and progression retinopathy remain unclear. OBJECTIVE TPL2 (tumor progression locus 2), a serine-threonine protein kinase, exerts a pathological effect on vascular angiogenesis. This study investigated the role of Nε-(carboxymethyl)lysine, a major advanced glycation end products, and the involved TPL2-related molecular signals in diabetic retinopathy using models of in vitro and in vivo and human samples. METHODS AND RESULTS Serum Nε-(carboxymethyl)lysine levels and TPL2 kinase activity were significantly increased in clinical patients and experimental animals with diabetic retinopathy. Intravitreal administration of pharmacological blocker or neutralizing antibody inhibited TPL2 and effectively suppressed the pathological characteristics of retinopathy in streptozotocin-induced diabetic animal models. Intravitreal VEGF (vascular endothelial growth factor) neutralization also suppressed the diabetic retinopathy in diabetic animal models. Mechanistic studies in primary human umbilical vein endothelial cells and primary retinal microvascular endothelial cells from streptozotocin-diabetic rats, db/db mice, and samples from patients with diabetic retinopathy revealed a positive parallel correlation between Nε-(carboxymethyl)lysine and the TPL2/chemokine SDF1α (stromal cell-derived factor-α) axis that is dependent on endoplasmic reticulum stress-related molecules, especially ATF4 (activating transcription factor-4). CONCLUSIONS This study demonstrates that inhibiting the Nε-(carboxymethyl)lysine-induced TPL2/ATF4/SDF1α axis can effectively prevent diabetes mellitus-mediated retinal microvascular dysfunction. This signaling axis may include the therapeutic potential for other diseases involving pathological neovascularization or macular edema.
Collapse
Affiliation(s)
- De-Wei Lai
- From the Institute of Biomedical Sciences (D.-W.L., M.-L.S.), Department of Chemistry (M.-R.L., C.-Y.C.), Rong Hsing Research Center for Translational Medicine (K.-H.L., W.H.-H.S., M.-L.S.), National Chung Hsing University, Taichung, Taiwan; Department of Ophthalmology (K.-H.L.), Division of Endocrinology and Metabolism (W.H.-H.S.), and Department of Medical Research (W.-J.L., Y.-W.H., M.-L.S.), Taichung Veterans General Hospital, Taiwan; Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan (C.-C.S.); Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (T.-J.C.); Department of Internal Medicine, Armed Forces Taichung General Hospital, Taiwan (T.-J.C.); Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (S.-H.L.); and Institute of Toxicology, College of Medicine, National Taiwan University, Taipei (S.-H.L.)
| | - Keng-Hung Lin
- From the Institute of Biomedical Sciences (D.-W.L., M.-L.S.), Department of Chemistry (M.-R.L., C.-Y.C.), Rong Hsing Research Center for Translational Medicine (K.-H.L., W.H.-H.S., M.-L.S.), National Chung Hsing University, Taichung, Taiwan; Department of Ophthalmology (K.-H.L.), Division of Endocrinology and Metabolism (W.H.-H.S.), and Department of Medical Research (W.-J.L., Y.-W.H., M.-L.S.), Taichung Veterans General Hospital, Taiwan; Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan (C.-C.S.); Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (T.-J.C.); Department of Internal Medicine, Armed Forces Taichung General Hospital, Taiwan (T.-J.C.); Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (S.-H.L.); and Institute of Toxicology, College of Medicine, National Taiwan University, Taipei (S.-H.L.)
| | - Wayne Huey-Herng Sheu
- From the Institute of Biomedical Sciences (D.-W.L., M.-L.S.), Department of Chemistry (M.-R.L., C.-Y.C.), Rong Hsing Research Center for Translational Medicine (K.-H.L., W.H.-H.S., M.-L.S.), National Chung Hsing University, Taichung, Taiwan; Department of Ophthalmology (K.-H.L.), Division of Endocrinology and Metabolism (W.H.-H.S.), and Department of Medical Research (W.-J.L., Y.-W.H., M.-L.S.), Taichung Veterans General Hospital, Taiwan; Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan (C.-C.S.); Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (T.-J.C.); Department of Internal Medicine, Armed Forces Taichung General Hospital, Taiwan (T.-J.C.); Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (S.-H.L.); and Institute of Toxicology, College of Medicine, National Taiwan University, Taipei (S.-H.L.)
| | - Maw-Rong Lee
- From the Institute of Biomedical Sciences (D.-W.L., M.-L.S.), Department of Chemistry (M.-R.L., C.-Y.C.), Rong Hsing Research Center for Translational Medicine (K.-H.L., W.H.-H.S., M.-L.S.), National Chung Hsing University, Taichung, Taiwan; Department of Ophthalmology (K.-H.L.), Division of Endocrinology and Metabolism (W.H.-H.S.), and Department of Medical Research (W.-J.L., Y.-W.H., M.-L.S.), Taichung Veterans General Hospital, Taiwan; Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan (C.-C.S.); Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (T.-J.C.); Department of Internal Medicine, Armed Forces Taichung General Hospital, Taiwan (T.-J.C.); Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (S.-H.L.); and Institute of Toxicology, College of Medicine, National Taiwan University, Taipei (S.-H.L.)
| | - Chung-Yu Chen
- From the Institute of Biomedical Sciences (D.-W.L., M.-L.S.), Department of Chemistry (M.-R.L., C.-Y.C.), Rong Hsing Research Center for Translational Medicine (K.-H.L., W.H.-H.S., M.-L.S.), National Chung Hsing University, Taichung, Taiwan; Department of Ophthalmology (K.-H.L.), Division of Endocrinology and Metabolism (W.H.-H.S.), and Department of Medical Research (W.-J.L., Y.-W.H., M.-L.S.), Taichung Veterans General Hospital, Taiwan; Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan (C.-C.S.); Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (T.-J.C.); Department of Internal Medicine, Armed Forces Taichung General Hospital, Taiwan (T.-J.C.); Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (S.-H.L.); and Institute of Toxicology, College of Medicine, National Taiwan University, Taipei (S.-H.L.)
| | - Wen-Jane Lee
- From the Institute of Biomedical Sciences (D.-W.L., M.-L.S.), Department of Chemistry (M.-R.L., C.-Y.C.), Rong Hsing Research Center for Translational Medicine (K.-H.L., W.H.-H.S., M.-L.S.), National Chung Hsing University, Taichung, Taiwan; Department of Ophthalmology (K.-H.L.), Division of Endocrinology and Metabolism (W.H.-H.S.), and Department of Medical Research (W.-J.L., Y.-W.H., M.-L.S.), Taichung Veterans General Hospital, Taiwan; Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan (C.-C.S.); Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (T.-J.C.); Department of Internal Medicine, Armed Forces Taichung General Hospital, Taiwan (T.-J.C.); Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (S.-H.L.); and Institute of Toxicology, College of Medicine, National Taiwan University, Taipei (S.-H.L.)
| | - Yi-Wen Hung
- From the Institute of Biomedical Sciences (D.-W.L., M.-L.S.), Department of Chemistry (M.-R.L., C.-Y.C.), Rong Hsing Research Center for Translational Medicine (K.-H.L., W.H.-H.S., M.-L.S.), National Chung Hsing University, Taichung, Taiwan; Department of Ophthalmology (K.-H.L.), Division of Endocrinology and Metabolism (W.H.-H.S.), and Department of Medical Research (W.-J.L., Y.-W.H., M.-L.S.), Taichung Veterans General Hospital, Taiwan; Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan (C.-C.S.); Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (T.-J.C.); Department of Internal Medicine, Armed Forces Taichung General Hospital, Taiwan (T.-J.C.); Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (S.-H.L.); and Institute of Toxicology, College of Medicine, National Taiwan University, Taipei (S.-H.L.)
| | - Chin-Chang Shen
- From the Institute of Biomedical Sciences (D.-W.L., M.-L.S.), Department of Chemistry (M.-R.L., C.-Y.C.), Rong Hsing Research Center for Translational Medicine (K.-H.L., W.H.-H.S., M.-L.S.), National Chung Hsing University, Taichung, Taiwan; Department of Ophthalmology (K.-H.L.), Division of Endocrinology and Metabolism (W.H.-H.S.), and Department of Medical Research (W.-J.L., Y.-W.H., M.-L.S.), Taichung Veterans General Hospital, Taiwan; Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan (C.-C.S.); Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (T.-J.C.); Department of Internal Medicine, Armed Forces Taichung General Hospital, Taiwan (T.-J.C.); Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (S.-H.L.); and Institute of Toxicology, College of Medicine, National Taiwan University, Taipei (S.-H.L.)
| | - Tsung-Ju Chung
- From the Institute of Biomedical Sciences (D.-W.L., M.-L.S.), Department of Chemistry (M.-R.L., C.-Y.C.), Rong Hsing Research Center for Translational Medicine (K.-H.L., W.H.-H.S., M.-L.S.), National Chung Hsing University, Taichung, Taiwan; Department of Ophthalmology (K.-H.L.), Division of Endocrinology and Metabolism (W.H.-H.S.), and Department of Medical Research (W.-J.L., Y.-W.H., M.-L.S.), Taichung Veterans General Hospital, Taiwan; Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan (C.-C.S.); Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (T.-J.C.); Department of Internal Medicine, Armed Forces Taichung General Hospital, Taiwan (T.-J.C.); Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (S.-H.L.); and Institute of Toxicology, College of Medicine, National Taiwan University, Taipei (S.-H.L.)
| | - Shing-Hwa Liu
- From the Institute of Biomedical Sciences (D.-W.L., M.-L.S.), Department of Chemistry (M.-R.L., C.-Y.C.), Rong Hsing Research Center for Translational Medicine (K.-H.L., W.H.-H.S., M.-L.S.), National Chung Hsing University, Taichung, Taiwan; Department of Ophthalmology (K.-H.L.), Division of Endocrinology and Metabolism (W.H.-H.S.), and Department of Medical Research (W.-J.L., Y.-W.H., M.-L.S.), Taichung Veterans General Hospital, Taiwan; Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan (C.-C.S.); Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (T.-J.C.); Department of Internal Medicine, Armed Forces Taichung General Hospital, Taiwan (T.-J.C.); Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (S.-H.L.); and Institute of Toxicology, College of Medicine, National Taiwan University, Taipei (S.-H.L.)
| | - Meei-Ling Sheu
- From the Institute of Biomedical Sciences (D.-W.L., M.-L.S.), Department of Chemistry (M.-R.L., C.-Y.C.), Rong Hsing Research Center for Translational Medicine (K.-H.L., W.H.-H.S., M.-L.S.), National Chung Hsing University, Taichung, Taiwan; Department of Ophthalmology (K.-H.L.), Division of Endocrinology and Metabolism (W.H.-H.S.), and Department of Medical Research (W.-J.L., Y.-W.H., M.-L.S.), Taichung Veterans General Hospital, Taiwan; Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan (C.-C.S.); Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (T.-J.C.); Department of Internal Medicine, Armed Forces Taichung General Hospital, Taiwan (T.-J.C.); Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (S.-H.L.); and Institute of Toxicology, College of Medicine, National Taiwan University, Taipei (S.-H.L.).
| |
Collapse
|
20
|
Shanahan CM, Furmanik M. Endoplasmic Reticulum Stress in Arterial Smooth Muscle Cells: A Novel Regulator of Vascular Disease. Curr Cardiol Rev 2017; 13:94-105. [PMID: 27758694 PMCID: PMC5440785 DOI: 10.2174/1573403x12666161014094738] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/24/2016] [Accepted: 10/06/2016] [Indexed: 01/27/2023] Open
Abstract
Cardiovascular disease continues to be the leading cause of death in industrialised societies. The idea that the arterial smooth muscle cell (ASMC) plays a key role in regulating many vascular pathologies has been gaining importance, as has the realisation that not enough is known about the pathological cellular mechanisms regulating ASMC function in vascular remodelling. In the past decade endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) have been recognised as a stress response underlying many physiological and pathological processes in various vascular cell types. Here we summarise what is known about how ER stress signalling regulates phenotypic switching, trans/dedifferentiation and apoptosis of ASMCs and contributes to atherosclerosis, hypertension, aneurysms and vascular calcification.
Collapse
Affiliation(s)
- Catherine M Shanahan
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, James Black Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, United Kingdom
| | - Malgorzata Furmanik
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, James Black Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, United Kingdom
| |
Collapse
|
21
|
Bodewes TCF, Johnson JM, Auster M, Huynh C, Muralidharan S, Contreras M, LoGerfo FW, Pradhan-Nabzdyk L. Intraluminal delivery of thrombospondin-2 small interfering RNA inhibits the vascular response to injury in a rat carotid balloon angioplasty model. FASEB J 2016; 31:109-119. [PMID: 27671229 DOI: 10.1096/fj.201600501r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 09/16/2016] [Indexed: 01/06/2023]
Abstract
In an effort to inhibit the response to vascular injury that leads to intimal hyperplasia, this study investigated the in vivo efficacy of intraluminal delivery of thrombospondin-2 (TSP-2) small interfering RNA (siRNA). Common carotid artery (CCA) balloon angioplasty injury was performed in rats. Immediately after denudation, CCA was transfected intraluminally (15 min) with one of the following: polyethylenimine (PEI)+TSP-2 siRNA, saline, PEI only, or PEI+control siRNA. CCA was analyzed at 24 h or 21 d by using quantitative real-time PCR and immunohistochemistry. TSP-2 gene and protein expression were significantly up-regulated after endothelial denudation at 24 h and 21 d compared with contralateral untreated, nondenuded CCA. Treatment with PEI+TSP-2 siRNA significantly suppressed TSP-2 gene expression (3.1-fold) at 24 h and TSP-2 protein expression, cell proliferation, and collagen deposition up to 21 d. These changes could be attributed to changes in TGF-β and matrix metalloproteinase-9, the downstream effectors of TSP-2. TSP-2 knockdown induced anti-inflammatory M2 macrophage polarization at 21 d; however, it did not significantly affect intima/media ratios. In summary, these data demonstrate effective siRNA transfection of the injured arterial wall and provide a clinically effective and translationally applicable therapeutic strategy that involves nonviral siRNA delivery to ameliorate the response to vascular injury.-Bodewes, T. C. F., Johnson, J. M., Auster, M., Huynh, C., Muralidharan, S., Contreras, M., LoGerfo, F. W., Pradhan-Nabzdyk, L. Intraluminal delivery of thrombospondin-2 small interfering RNA inhibits the vascular response to injury in a rat carotid balloon angioplasty model.
Collapse
Affiliation(s)
- Thomas C F Bodewes
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Vascular Surgery, University Medical Center, Utrecht, The Netherlands; and
| | - Joel M Johnson
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Auster
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Cindy Huynh
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Surgery, State University of New York (SUNY) Upstate Medical University, Syracuse, New York, USA
| | - Sriya Muralidharan
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mauricio Contreras
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Frank W LoGerfo
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Leena Pradhan-Nabzdyk
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
22
|
Kovaleva IE, Garaeva AA, Chumakov PM, Evstafieva AG. Intermedin/adrenomedullin 2 is a stress-inducible gene controlled by activating transcription factor 4. Gene 2016; 590:177-85. [DOI: 10.1016/j.gene.2016.06.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 06/03/2016] [Accepted: 06/17/2016] [Indexed: 10/21/2022]
|
23
|
Friend K, Brooks HA, Propson NE, Thomson JA, Kimble J. Embryonic Stem Cell Growth Factors Regulate eIF2α Phosphorylation. PLoS One 2015; 10:e0139076. [PMID: 26406898 PMCID: PMC4583406 DOI: 10.1371/journal.pone.0139076] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/09/2015] [Indexed: 12/18/2022] Open
Abstract
Growth factors and transcription factors are well known to regulate pluripotent stem cells, but less is known about translational control in stem cells. Here, we use embryonic stem cells (ESCs) to investigate a connection between ESC growth factors and eIF2α-mediated translational control (eIF2α phosphorylation promotes protein expression from mRNAs with upstream open-reading frames, or uORFs). We find abundant phosphorylated P-eIF2α (P-eIF2α) in both pluripotent mouse and human ESCs, but little P-eIF2α in ESCs triggered to differentiate. We show that the growth factors LIF (leukemia inhibitory factor) and BMP4 (bone morphogenic protein 4) both maintain P-eIF2α in mESCs, but use distinct mechanisms: LIF inhibits an eIF2α phosphatase whereas BMP4 activates an eIF2α kinase. The mRNAs encoding the pluripotency factors Nanog and c-Myc possess uORFs while Oct4 mRNA does not. We find that salubrinal, a chemical that increases eIF2α phosphorylation, promotes Nanog and c-Myc expression, but not Oct4 expression. These experiments connect ESC growth factors to eIF2α phosphorylation and suggest a chemical substitute for LIF to enhance Nanog and c-Myc expression.
Collapse
Affiliation(s)
- Kyle Friend
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
- Department of Chemistry and Biochemistry, Washington and Lee University, Lexington, Virginia, 24450, United States of America
- * E-mail: (KF); (JK)
| | - Hunter A. Brooks
- Department of Chemistry and Biochemistry, Washington and Lee University, Lexington, Virginia, 24450, United States of America
| | - Nicholas E. Propson
- The Morgridge Institute for Research, 309 North Orchard Street, Madison, Wisconsin, 53715, United States of America
| | - James A. Thomson
- The Morgridge Institute for Research, 309 North Orchard Street, Madison, Wisconsin, 53715, United States of America
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706, United States of America
| | - Judith Kimble
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
- * E-mail: (KF); (JK)
| |
Collapse
|
24
|
Xu F, Ahmed ASI, Kang X, Hu G, Liu F, Zhang W, Zhou J. MicroRNA-15b/16 Attenuates Vascular Neointima Formation by Promoting the Contractile Phenotype of Vascular Smooth Muscle Through Targeting YAP. Arterioscler Thromb Vasc Biol 2015; 35:2145-52. [PMID: 26293467 DOI: 10.1161/atvbaha.115.305748] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/10/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To investigate the functional role of the microRNA (miR)-15b/16 in vascular smooth muscle (SM) phenotypic modulation. APPROACH AND RESULTS We found that miR-15b/16 is one of the most abundant mRs expressed in contractile vascular smooth muscle cells (VSMCs). However, when contractile VSMCs get converted to a synthetic phenotype, miR-15b/16 expression is significantly reduced. Knocking down endogenous miR-15b/16 in VSMCs attenuates SM-specific gene expression but promotes VSMC proliferation and migration. Conversely, overexpression of miR-15b/16 promotes SM contractile gene expression while attenuating VSMC migration and proliferation. Consistent with this, overexpression of miR-15b/16 in a rat carotid balloon injury model markedly attenuates injury-induced SM dedifferentiation and neointima formation. Mechanistically, we identified the potent oncoprotein yes-associated protein (YAP) as a downstream target of miR-15b/16 in VSMCs. Reporter assays validated that miR-15b/16 targets YAP's 3' untranslated region. Moreover, overexpression of miR-15b/16 significantly represses YAP expression, whereas conversely, depletion of endogenous miR-15b/16 results in upregulation of YAP expression. CONCLUSIONS These results indicate that miR-15b/16 plays a critical role in SM phenotypic modulation at least partly through targeting YAP. Restoring expression of miR-15b/16 would be a potential therapeutic approach for treatment of proliferative vascular diseases.
Collapse
Affiliation(s)
- Fei Xu
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Abu Shufian Ishtiaq Ahmed
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Xiuhua Kang
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Guoqing Hu
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Fang Liu
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Wei Zhang
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Jiliang Zhou
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.).
| |
Collapse
|
25
|
McDonald AI, Iruela-Arispe ML. Healing arterial ulcers: Endothelial lining regeneration upon vascular denudation injury. Vascul Pharmacol 2015; 72:9-15. [PMID: 26093336 DOI: 10.1016/j.vph.2015.06.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 06/16/2015] [Indexed: 12/20/2022]
Abstract
Thrombosis and restenosis are the most prevalent late complications of coronary artery stenting. Current standards of clinical care focus on prevention of smooth muscle cell proliferation by the use of drug-eluting stents able to release anti-proliferative drugs. Unfortunately, these drugs also block endothelial cell proliferation and, in this manner, prevent recovery of endothelial cell coverage. Continued lack of endothelial repair leaves the root cause of thrombosis and restenosis unchanged, creating a vicious cycle where drug-mediated prevention of restenosis simultaneously implies promotion of thrombosis. In this issue of Vascular Pharmacology, Hussner and colleagues provide in vitro evidence and a mechanistic basis for the use of atorvastatin in stents as a way to bypass this roadblock. Here we review the pathological mechanisms and therapeutic approaches to restore flow in occluded arteries. We argue that rational design of drug eluting stents should focus on specific inhibition of smooth muscle cell proliferation with concurrent stimulation of endothelial regeneration. We comment on the current poor understanding of the cellular and molecular regulation of endothelial cell proliferation in the context of a functional artery, and on the pitfalls of extrapolating from the well-studied process of neovascularization by sprouting vessel formation.
Collapse
Affiliation(s)
- Austin I McDonald
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - M Luisa Iruela-Arispe
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA..
| |
Collapse
|
26
|
Impact of ellagic acid in bone formation after tooth extraction: an experimental study on diabetic rats. ScientificWorldJournal 2014; 2014:908098. [PMID: 25485304 PMCID: PMC4251085 DOI: 10.1155/2014/908098] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 08/05/2014] [Accepted: 08/26/2014] [Indexed: 01/10/2023] Open
Abstract
Objectives. To estimate the impact of ellagic acid (EA) towards healing tooth socket in diabetic animals, after tooth extraction. Methods. Twenty-four Sprague Dawley male rats weighing 250–300 g were selected for this study. All animals were intraperitoneally injected with 45 mg/kg (b.w.) of freshly prepared streptozotocin (STZ), to induce diabetic mellitus. Then, the animals were anesthetized, and the upper left central incisor was extracted and the whole extracted sockets were filled with Rosuvastatin (RSV). The rats were separated into three groups, comprising 8 rats each. The first group was considered as normal control group and orally treated with normal saline. The second group was regarded as diabetic control group and orally treated with normal saline, whereas the third group comprised diabetic rats, administrated with EA (50 mg/kg) orally. The maxilla tissue stained by eosin and hematoxylin (H&E) was used for histological examinations and immunohistochemical technique. Fibroblast growth factor (FGF-2) and alkaline phosphatase (ALP) were used to evaluate the healing process in the extracted tooth socket by immunohistochemistry test. Results. The reactions of immunohistochemistry for FGF-2 and ALP presented stronger expression, predominantly in EA treated diabetic rat, than the untreated diabetic rat. Conclusion. These findings suggest that the administration of EA combined with RSV may have accelerated the healing process of the tooth socket of diabetic rats, after tooth extraction.
Collapse
|
27
|
Ran F, Liu C, Liu Z, Shang T, Zhou M, Qiao T. Preventive effects of basic fibroblast growth factor on vascular restenosis after balloon angioplasty. Exp Ther Med 2014; 7:1193-1196. [PMID: 24940410 PMCID: PMC3991498 DOI: 10.3892/etm.2014.1562] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 01/29/2014] [Indexed: 12/28/2022] Open
Abstract
The aim of the present study was to investigate whether chronic administration of basic fibroblast growth factor (bFGF) following angioplasty in a dog model of atherosclerotic iliac stenosis may restore endothelium function and prevent restenosis (RS). In total, 40 dogs with atherosclerotic stenosis of the right iliac arteries were used in the study. A total of 20 dogs underwent histological examination of the lumen areas prior to (n=10) and immediately following angioplasty (n=10). Intravenous bFGF was administered to 10 dogs (bFGF group) and an additional 10 dogs received vehicle injection (control group). Animals in the two groups were sacrificed 42 days following surgery for in vitro analysis of vascular reactivity and morphometric assessment of the histological cross-sectional areas. The bFGF group exhibited significantly greater maximal endothelium-dependent acetylcholine-induced relaxation (Emax, 43±9%) when compared with the control group (Emax, 8±6%; P<0.05). In addition, the maximal endothelium-independent response of the bFGF group to sodium nitroprusside (Emax, 90±2%) was greater than that of the control group (Emax, 60±2%; P<0.05). Six weeks following angioplasty, the lumen area in the bFGF group (2.01±0.78 mm2) was greater compared with the control group (1.0±0.10%). The lumen area decreased by 58% between immediately after angioplasty and the control group six weeks following angioplasty. Therefore, the results of the present study indicated that administration of bFGF may not only restore endothelium-dependent and -independent relaxation, but also prevent RS in dogs that have undergone angioplasty.
Collapse
Affiliation(s)
- Feng Ran
- Department of Vascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Changjian Liu
- Department of Vascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Zhao Liu
- Department of Vascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Tao Shang
- Department of Vascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Min Zhou
- Department of Vascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Tong Qiao
- Department of Vascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
28
|
Ishii T, Mann GE. Redox status in mammalian cells and stem cells during culture in vitro: critical roles of Nrf2 and cystine transporter activity in the maintenance of redox balance. Redox Biol 2014; 2:786-94. [PMID: 25009780 PMCID: PMC4085355 DOI: 10.1016/j.redox.2014.04.008] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 04/14/2014] [Accepted: 04/15/2014] [Indexed: 12/24/2022] Open
Abstract
Culturing cells and tissues in vitro has provided valuable insights into the molecular mechanisms regulating redox signaling in cells with implications for medicine. However, standard culture techniques maintain mammalian cells in vitro under an artificial physicochemical environment such as ambient air and 5% CO2. Oxidative stress is caused by the rapid oxidation of cysteine to cystine in culture media catalyzed by transition metals, leading to diminished intracellular cysteine and glutathione (GSH) pools. Some cells, such as fibroblasts and macrophages, express cystine transport activity, designated as system [Formula: see text], which enables cells to maintain these pools to counteract oxidative stress. Additionally, many cells have the ability to activate the redox sensitive transcription factor Nrf2, a master regulator of cellular defenses against oxidative stress, and to upregulate xCT, the subunit of the [Formula: see text] transport system leading to increases in cellular GSH. In contrast, some cells, including lymphoid cells, embryonic stem cells and iPS cells, express relatively low levels of xCT and cannot maintain cellular cysteine and GSH pools. Thus, fibroblasts have been used as feeder cells for the latter cell types based on their ability to supply cysteine. Other key Nrf2 regulated gene products include heme oxygenase 1, peroxiredoxin 1 and sequestosome1. In macrophages, oxidized LDL activates Nrf2 and upregulates the scavenger receptor CD36 forming a positive feedback loop to facilitate removal of the oxidant from the vascular microenvironment. This review describes cell type specific responses to oxygen derived stress, and the key roles that activation of Nrf2 and membrane transport of cystine and cysteine play in the maintenance and proliferation of mammalian cells in culture.
Collapse
Key Words
- 2-Mercaptoethanol
- 4HNE, 4-hydroxynonenal
- BCS, bathocuproine sulfonate
- CD36
- Cystine transporter
- ES cells, embryonic stem cells
- Embryonic stem cells
- Feeder cells
- Glutathione
- HO-1, heme oxygenase 1
- Keap1, Kelch-like ECH-associated protein 1
- Lymphocytes
- MRPs, multidrug resistance-associated proteins
- Nrf2
- Nrf2, nuclear factor erythroid 2-related factor 2
- Oxygen
- Prx1, peroxiredoxin 1
- SQSTM1, sequestosome1
- iPS cells
- iPS cells, induced pluripotent stem cells
- oxLDL, oxidized low density lipoprotein
- xCT
Collapse
Affiliation(s)
- Tetsuro Ishii
- University of Tsukuba, Ibaraki, Japan
- Corresponding author:
| | - Giovanni E. Mann
- Cardiovascular Division, British Heart Foundation Centre of Research Excellence, School of Medicine, King's College London, 150 Stamford Street, London SE1 9NH, UK
| |
Collapse
|
29
|
Li Y, Bhindi R, Deng ZJ, Morton SW, Hammond PT, Khachigian LM. Inhibition of vein graft stenosis with a c-jun targeting DNAzyme in a cationic liposomal formulation containing 1,2-dioleoyl-3-trimethylammonium propane (DOTAP)/1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE). Int J Cardiol 2013; 168:3659-64. [PMID: 23886527 PMCID: PMC3951723 DOI: 10.1016/j.ijcard.2013.05.092] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 05/02/2013] [Accepted: 05/31/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES Coronary artery bypass grafting (CABG) is among the most commonly performed heart surgical procedures. Saphenous vein graft failure due to stenosis impedes the longer-term success of CABG. A key cellular event in the process of vein graft stenosis is smooth muscle cell hyperplasia. In this study, we evaluated the effect of a DNAzyme (Dz13) targeting the transcription factor c-Jun in a rabbit model of vein graft stenosis in a cationic liposomal formulation containing 1,2-dioleoyl-3-trimethylammonium propane (DOTAP)/1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE). Dz13 in DOTAP/DOPE has undergone preclinical toxicological testing, and a Phase I clinical trial we recently conducted in basal cell carcinoma cancer patients demonstrates that it is safe and well tolerated after local administration. METHODS Effects of Dz13 in a formulation containing DOTAP/DOPE on smooth muscle cell (SMC) growth and c-Jun expression were assessed. Dz13 transfection was determined by cellular uptake of carboxyfluorescein-labeled Dz13. Autologous jugular vein to carotid artery transplantation was performed in New Zealand White rabbits to investigate the effect of the Dz13 in DOTAP/DOPE formulation on intimal hyperplasia. RESULTS Dz13/DOTAP/DOPE reduced SMC proliferation and c-Jun protein expression in vitro compared with an impotent form of Dz13 bearing a point mutation in its catalytic domain (Dz13.G>C). The Dz13(500 μg)/DOTAP/DOPE formed lipoplexes that were colloidally stable for up to 1h on ice (0°C) and 30 min at 37°C, allowing sufficient uptake by the veins. Dz13 (500 μg) inhibited neointima formation 28 d after end-to-side transplantation. CONCLUSIONS This formulation applied to veins prior to transplantation may potentially be useful in efforts to reduce graft failure.
Collapse
Affiliation(s)
- Yue Li
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | | | | | | | | | | |
Collapse
|
30
|
Xiang J, Zhao Y, Chen J, Zhou J. Expression of basic fibroblast growth factor, protein kinase C and members of the apoptotic pathway in skeletal muscle of streptozotocin-induced diabetic rats. Tissue Cell 2013; 46:1-8. [PMID: 24008114 DOI: 10.1016/j.tice.2013.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/22/2013] [Accepted: 07/22/2013] [Indexed: 01/11/2023]
Abstract
This study investigated the potential mechanisms that may underlie diabetes induced amyoatrophy. Sprague-Dawley rats were either injected intraperiotneally with STZ (test group; N=8) to induce diabetic-like symptoms (blood glucose level ≥16.65mmol/L) or with buffer (control group; N=8). Differences in muscle structure between the STZ-induced diabetic and control groups were evaluated by histochemistry. Protein and mRNA levels of basic FGF (bFGF), bax, bcl-2, and caspase 3 in skeletal muscle were compared between the 2 groups using immunohistochemistry and quantitative PCR, respectively. Serum level of insulin and protein kinase C (PKC) were measured by competitive RIA and ELISA, respectively. Unlike control animals, the skeletal muscle fibers from STZ-induced diabetic animals were broken and pyknotic, the sarcomeric structure disrupted, and mild hyperplasia of interstitial adipose tissues was detected. The serum level of PKC was higher (P=0.003) and the protein and mRNA levels of bFGF in skeletal muscle were lower (P=0.001) in STZ-induced diabetic versus control animals. Protein and mRNA levels of the apoptosis promoting genes caspase-3 and bax were higher in skeletal muscle from STZ-induced diabetic rats as compared to control animals (P<0.001 and P=0.037, respectively), while mRNA and protein levels of bcl-2, an inhibitor of apoptosis, was lower in STZ-induced diabetic rats versus control animals (P=0.026). Increasing apoptosis in skeletal muscle from STZ-induced diabetic rats was further demonstrated by TNNEL assay. Our findings suggest that enhanced PKC levels, reduction of bFGF expression, and increased in apoptosis might be associated with the development of diabetes-induced myoatrophy.
Collapse
Affiliation(s)
- Jingyan Xiang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yuwu Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Jingjiong Chen
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jian Zhou
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
31
|
Chai L, Ling K, He X, Yang R. Expression of ATF4 and VEGF in chorionic villus tissue in early spontaneous abortion. Eur J Obstet Gynecol Reprod Biol 2013; 170:434-8. [PMID: 23891064 DOI: 10.1016/j.ejogrb.2013.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Revised: 04/18/2013] [Accepted: 07/02/2013] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To explore the relationship between early spontaneous abortion (SA) and the expression of activating transcription factor 4 (ATF4) and vascular endothelial growth factor (VEGF). STUDY DESIGN The expression of ATF4 and VEGF protein and mRNA in villi from first trimester spontaneous abortion (SA, n=30) and normal pregnancy (NP, n=30) were detected by immunohistochemistry and fluorescent quantitative polymerase chain reaction (FQ-PCR). RESULTS Both protein and mRNA expressions of ATF4 and VEGF in the SA group were significantly lower than in the NP group (P<0.01). Their proteins are expressed mainly in syncytiotrophoblast, cytotrophoblast and villous stromal cells. Correlation analysis showed that the expression of ATF4 was positively correlated with that of VEGF in the SA group (r=0.717, P<0.01). CONCLUSION Lower expression of ATF4 and VEGF genes in chorionic villus tissue may participate in the pathogenesis of spontaneous abortion.
Collapse
Affiliation(s)
- Luwei Chai
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | | | | | | |
Collapse
|
32
|
HLA-B35 and dsRNA induce endothelin-1 via activation of ATF4 in human microvascular endothelial cells. PLoS One 2013; 8:e56123. [PMID: 23441162 PMCID: PMC3575387 DOI: 10.1371/journal.pone.0056123] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 01/05/2013] [Indexed: 12/17/2022] Open
Abstract
Endothelin 1 (ET-1) is a key regulator of vascular homeostasis. We have recently reported that the presence of Human antigen class I, HLA-B35, contributes to human dermal microvascular endothelial cell (HDMEC) dysfunction by upregulating ET-1 and proinflammatory genes. Likewise, a Toll-like receptor 3 (TLR3) ligand, Poly(I:C), was shown to induce ET-1 expression in HDMECs. The goal of this study was to determine the molecular mechanism of ET-1 induction by these two agonists. Because HLA-B35 expression correlated with induction of Binding Immunoglobulin Protein (BiP/GRP78) and several heat shock proteins, we first focused on ER stress and unfolded protein response (UPR) as possible mediators of this response. ER stress inducer, Thapsigargin (TG), HLA-B35, and Poly(I:C) induced ET-1 expression with similar potency in HDMECs. TG and HLA-B35 activated the PERK/eIF2α/ATF4 branch of the UPR and modestly increased the spliced variant of XBP1, but did not affect the ATF6 pathway. Poly(I:C) also activated eIF2α/ATF4 in a protein kinase R (PKR)-dependent manner. Depletion of ATF4 decreased basal expression levels of ET-1 mRNA and protein, and completely prevented upregulation of ET-1 by all three agonists. Additional experiments have demonstrated that the JNK and NF-κB pathways are also required for ET-1 upregulation by these agonists. Formation of the ATF4/c-JUN complex, but not the ATF4/NF-κB complex was increased in the agonist treated cells. The functional role of c-JUN in responses to HLA-B35 and Poly(I:C) was further confirmed in ET-1 promoter assays. This study identified ATF4 as a novel activator of the ET-1 gene. The ER stress/UPR and TLR3 pathways converge on eIF2α/ATF4 during activation of endothelial cells.
Collapse
|
33
|
Afonyushkin T, Oskolkova OV, Bochkov VN. Permissive role of miR-663 in induction of VEGF and activation of the ATF4 branch of unfolded protein response in endothelial cells by oxidized phospholipids. Atherosclerosis 2012; 225:50-5. [PMID: 22776647 DOI: 10.1016/j.atherosclerosis.2012.06.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 05/25/2012] [Accepted: 06/09/2012] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Atherosclerotic lesions contain high concentrations of oxidized phospholipids (OxPLs) known to induce VEGF via the ATF4 arm of unfolded protein response (UPR), and to promote angiogenic reactions thus potentially contributing to the progression and destabilization of atherosclerotic plaques. In order to get further insights into the mechanisms of cellular stress-induced angiogenesis we studied the role of a specific microRNA (miR-663) in the mechanisms of VEGF induction by OxPLs and inducers of UPR. METHODS miRNA and mRNA levels were determined using microarray profiling and qRT-PCR methods. Proteins were analyzed by Western blotting. miR-663 levels were changed by transfecting cells with synthetic oligonucleotides. RESULTS OxPAPC elevated miR-663 in two types of human endothelial cells (ECs). Knockdown of miR-663 inhibited upregulation of VEGF mRNA in ECs treated by OxPAPC, OxPAPS or OxPAPA. In addition, silencing of miR-663 suppressed upregulation by OxPAPC of ATF4 mRNA and protein, as well as a downstream gene TRIB. Similarly to the inhibition of OxPAPC effects, knockdown of miR-663 suppressed elevation of ATF4, VEGF and TRIB in response to another inducer of UPR, tunicamycin. Overexpression of miR-663 reversed the inhibition of VEGF induction by miR-663 inhibitor. CONCLUSION miR-663 is critically important for 2 key events induced in ECs by stress agents and oxidized lipids, namely induction of transcription factor ATF4 and its downstream gene VEGF. These findings allow hypothesizing that miR-663 plays a general role in control of the ATF4 branch of UPR induced by different agents.
Collapse
Affiliation(s)
- Taras Afonyushkin
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, A-1090 Vienna, Austria
| | | | | |
Collapse
|
34
|
Malabanan KP, Sheahan AV, Khachigian LM. Platelet-derived growth factor-BB mediates cell migration through induction of activating transcription factor 4 and tenascin-C. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:2590-7. [PMID: 22507839 DOI: 10.1016/j.ajpath.2012.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2011] [Revised: 02/07/2012] [Accepted: 02/28/2012] [Indexed: 11/24/2022]
Abstract
The acute response to vascular cell injury, which underpins vasculo-occlusive pathologies such as atherogenesis and restenosis after percutaneous coronary intervention, involves a complex series of molecular events that alter patterns of gene expression and favor a synthetic phenotype. One transcription factor that has been implicated in this process is the evolutionarily conserved mammalian stress response pathway regulator activating transcription factor 4 (ATF-4). Here, we show for the first time that both mRNA and protein levels of ATF-4 are induced in smooth muscle cells (SMCs) by the potent migratory factor PDGF-BB through PDGFR-β. PDGF-BB also stimulates the expression of tenascin-C (TN-C), an extracellular matrix glycoprotein that regulates the activity of focal adhesion complexes, facilitating the SMC migration that underlies negative vascular remodeling in response to injury. Overexpression of ATF-4 increased transcript levels of the four TN-C isoforms in rat vascular SMCs, and ATF-4 knockdown inhibited PDGF-BB-inducible TN-C expression in vitro and injury-inducible TN-C protein expression in the balloon-injured rat artery wall. Furthermore, we show that ATF-4 is required for PDGF-BB-inducible SMC migration in response to injury. PDGF-BB-induced migration was also compromised in ATF-4 null mEFs, and this effect was rescued by the addition of TN-C. Our findings thus demonstrate the role of ATF-4 in both injury- and PDGF-BB-inducible TN-C expression and cell migration.
Collapse
|
35
|
Zhong Y, Li J, Chen Y, Wang JJ, Ratan R, Zhang SX. Activation of endoplasmic reticulum stress by hyperglycemia is essential for Müller cell-derived inflammatory cytokine production in diabetes. Diabetes 2012; 61:492-504. [PMID: 22228718 PMCID: PMC3266398 DOI: 10.2337/db11-0315] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Inflammation plays an important role in diabetes-induced retinal vascular leakage. The purpose of this study is to examine the role of endoplasmic reticulum (ER) stress and the signaling pathway of ER stress-induced activating transcription factor 4 (ATF4) in the regulation of Müller cell-derived inflammatory mediators in diabetic retinopathy. In diabetic animals, elevated ER stress markers, ATF4, and vascular endothelial growth factor (VEGF) expression were partially localized to Müller cells in the retina. In cultured Müller cells, high glucose induced a time-dependent increase of ER stress, ATF4 expression, and inflammatory factor production. Inducing ER stress or overexpressing ATF4 resulted in elevated intracellular adhesion molecule 1 and VEGF proteins in Müller cells. In contrast, alleviation of ER stress or blockade of ATF4 activity attenuated inflammatory gene expression induced by high glucose or hypoxia. Furthermore, we found that ATF4 regulated the c-Jun NH2-terminal kinase pathway resulting in VEGF upregulation. ATF4 was also required for ER stress-induced and hypoxia-inducible factor-1α activation. Finally, we showed that administration of chemical chaperone 4-phenylbutyrate or genetic inhibition of ATF4 successfully attenuated retinal VEGF expression and reduced vascular leakage in mice with STZ-induced diabetes. Taken together, our data indicate that ER stress and ATF4 play a critical role in retinal inflammatory signaling and Müller cell-derived inflammatory cytokine production in diabetes.
Collapse
Affiliation(s)
- Yimin Zhong
- Department of Medicine, Endocrinology, and Diabetes, Harold Hamm Oklahoma Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jingming Li
- Department of Medicine, Endocrinology, and Diabetes, Harold Hamm Oklahoma Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Yanming Chen
- Department of Medicine, Endocrinology, and Diabetes, Harold Hamm Oklahoma Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Medicine, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Joshua J. Wang
- Department of Medicine, Endocrinology, and Diabetes, Harold Hamm Oklahoma Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Rajiv Ratan
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, Burke Medical Research Institute, White Plains, New York
| | - Sarah X. Zhang
- Department of Medicine, Endocrinology, and Diabetes, Harold Hamm Oklahoma Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Corresponding author: Sarah X. Zhang,
| |
Collapse
|
36
|
Zhang SX, Sanders E, Wang JJ. Endoplasmic reticulum stress and inflammation: mechanisms and implications in diabetic retinopathy. J Ocul Biol Dis Infor 2012; 4:51-61. [PMID: 23330021 DOI: 10.1007/s12177-011-9075-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 12/29/2011] [Indexed: 12/21/2022] Open
Abstract
The endoplasmic reticulum (ER) is the primary cellular compartment where proteins are synthesized and modified before they can be transported to their destination. Dysfunction of the ER impairs protein homeostasis and leads to the accumulation of misfolded/unfolded proteins in the ER, or ER stress. While it has long been recognized that ER stress is a major cause of conformational disorders, such as Alzheimer's disease, Huntington's disease, certain types of cancer, and type 2 diabetes, recent evidence suggests that ER stress is also implicated in many chronic inflammatory diseases. These diseases include irritable bowel syndrome, atherosclerosis, diabetic complications, and many others. Diabetic retinopathy is a common microvascular complication of diabetes, characterized by chronic inflammation, progressive damage to retinal vascular and neuronal cells, vascular leakage, and abnormal blood vessel growth (neovascularization). In this review, we discuss the role and mechanisms of ER stress in retinal inflammation and vascular damage in diabetic retinopathy.
Collapse
Affiliation(s)
- Sarah X Zhang
- Department of Medicine, Endocrinology and Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA ; Harold Hamm Diabetes Center at University of Oklahoma, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA ; Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, 941 Stanton L. Young Blvd, Oklahoma City, OK USA
| | | | | |
Collapse
|
37
|
Activation transcription factor-4 and the acute vascular response to injury. J Mol Med (Berl) 2010; 88:545-52. [DOI: 10.1007/s00109-010-0615-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 02/14/2010] [Accepted: 02/25/2010] [Indexed: 11/25/2022]
|
38
|
Shan J, Lopez MC, Baker HV, Kilberg MS. Expression profiling after activation of amino acid deprivation response in HepG2 human hepatoma cells. Physiol Genomics 2010; 41:315-27. [PMID: 20215415 DOI: 10.1152/physiolgenomics.00217.2009] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Dietary protein malnutrition is manifested as amino acid deprivation of individual cells, which activates an amino acid response (AAR) that alters cellular functions, in part, by regulating transcriptional and posttranscriptional mechanisms. The AAR was activated in HepG2 human hepatoma cells, and the changes in mRNA content were analyzed by microarray expression profiling. The results documented that 1,507 genes were differentially regulated by P < 0.001 and by more than twofold in response to the AAR, 250 downregulated and 1,257 upregulated. The spectrum of altered genes reveals that amino acid deprivation has far-reaching implications for gene expression and cellular function. Among those cellular functions with the largest numbers of altered genes were cell growth and proliferation, cell cycle, gene expression, cell death, and development. Potential biological relationships between the differentially expressed genes were analyzed by computer software that generates gene networks. Proteins that were central to the most significant of these networks included c-myc, polycomb group proteins, transforming growth factor β1, nuclear factor (erythroid-derived 2)-like 2-related factor 2, FOS/JUN family members, and many members of the basic leucine zipper superfamily of transcription factors. Although most of these networks contained some genes that were known to be amino acid responsive, many new relationships were identified that underscored the broad impact that amino acid stress has on cellular function.
Collapse
Affiliation(s)
- Jixiu Shan
- Department of Biochemistry and Molecular Biology and
| | | | | | | |
Collapse
|
39
|
Afonyushkin T, Oskolkova OV, Philippova M, Resink TJ, Erne P, Binder BR, Bochkov VN. Oxidized phospholipids regulate expression of ATF4 and VEGF in endothelial cells via NRF2-dependent mechanism: novel point of convergence between electrophilic and unfolded protein stress pathways. Arterioscler Thromb Vasc Biol 2010; 30:1007-13. [PMID: 20185790 DOI: 10.1161/atvbaha.110.204354] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE The ATF4 arm of the unfolded protein response is increasingly recognized for its relevance to pathology, and in particular to angiogenic reactions. Oxidized phospholipids (OxPLs), known to accumulate in atherosclerotic vessels, were shown to upregulate vascular endothelial growth factor (VEGF) and induce angiogenesis via an ATF4-dependent mechanism. In this study, we analyzed the mechanism of ATF4 upregulation by OxPLs and more specifically the involvement of NRF2, the major transcriptional mediator of electrophilic stress response. METHODS AND RESULTS Using reverse transcription/real-time polymerase chain reaction and Western blotting, we found that OxPLs induced upregulation of ATF4 mRNA and protein in several types of endothelial cells and that these effects were suppressed by short interfering RNA (siRNA) against NRF2. Electrophilic (iso)prostaglandins and oxidized low-density lipoprotein, similarly to OxPLs, elevated ATF4 mRNA levels in an NRF2-dependent mode. Chromatin immunoprecipitation revealed OxPL-dependent binding of NRF2 to a putative antioxidant response element site in the ATF4 gene promoter. Knockdown of NRF2 inhibited OxPL-induced elevation of VEGF mRNA and endothelial cell sprout formation. CONCLUSION Our data characterize NRF2 as a positive regulator of ATF4 and identify a novel cross-talk between electrophilic and unfolded protein responses, which may play a role in stress-induced angiogenesis.
Collapse
Affiliation(s)
- Taras Afonyushkin
- Department of Vascular Biology and Thrombosis Research, Center for Biomolecular Medicine and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
40
|
Bisaillon JM, Motiani RK, Gonzalez-Cobos JC, Potier M, Halligan KE, Alzawahra WF, Barroso M, Singer HA, Jourd'heuil D, Trebak M. Essential role for STIM1/Orai1-mediated calcium influx in PDGF-induced smooth muscle migration. Am J Physiol Cell Physiol 2010; 298:C993-1005. [PMID: 20107038 DOI: 10.1152/ajpcell.00325.2009] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We recently demonstrated that thapsigargin-induced passive store depletion activates Ca(2+) entry in vascular smooth muscle cells (VSMC) through stromal interaction molecule 1 (STIM1)/Orai1, independently of transient receptor potential canonical (TRPC) channels. However, under physiological stimulations, despite the ubiquitous depletion of inositol 1,4,5-trisphosphate-sensitive stores, many VSMC PLC-coupled agonists (e.g., vasopressin and endothelin) activate various store-independent Ca(2+) entry channels. Platelet-derived growth factor (PDGF) is an important VSMC promigratory agonist with an established role in vascular disease. Nevertheless, the molecular identity of the Ca(2+) channels activated by PDGF in VSMC remains unknown. Here we show that inhibitors of store-operated Ca(2+) entry (Gd(3+) and 2-aminoethoxydiphenyl borate at concentrations as low as 5 microM) prevent PDGF-mediated Ca(2+) entry in cultured rat aortic VSMC. Protein knockdown of STIM1, Orai1, and PDGF receptor-beta (PDGFRbeta) impaired PDGF-mediated Ca(2+) influx, whereas Orai2, Orai3, TRPC1, TRPC4, and TRPC6 knockdown had no effect. Scratch wound assay showed that knockdown of STIM1, Orai1, or PDGFRbeta inhibited PDGF-mediated VSMC migration, but knockdown of STIM2, Orai2, and Orai3 was without effect. STIM1, Orai1, and PDGFRbeta mRNA levels were upregulated in vivo in VSMC from balloon-injured rat carotid arteries compared with noninjured control vessels. Protein levels of STIM1 and Orai1 were also upregulated in medial and neointimal VSMC from injured carotid arteries compared with noninjured vessels, as assessed by immunofluorescence microscopy. These results establish that STIM1 and Orai1 are important components for PDGF-mediated Ca(2+) entry and migration in VSMC and are upregulated in vivo during vascular injury and provide insights linking PDGF to STIM1/Orai1 during neointima formation.
Collapse
|
41
|
Fibroblast growth factor 2 positively regulates expression of activating transcription factor 4 in osteoblasts. Biochem Biophys Res Commun 2009; 391:335-9. [PMID: 19913500 DOI: 10.1016/j.bbrc.2009.11.059] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Accepted: 11/09/2009] [Indexed: 11/23/2022]
Abstract
Our previous studies showed that basic fibroblast growth factor 2 (FGF2) null mice display markedly reduced bone mass and bone formation. However, the mechanism by which FGF2 regulates bone mass or bone formation is not fully defined. Activating transcription factor 4 (ATF4), one member of activating transcription factor/cAMP response element binding family, is a transcription factor required for osteoblast terminal differentiation. Here we investigate the ability of FGF2 to increase expression of ATF4 in bone marrow stromal cells (BMSCs) and examine ATF4 expression in Fgf2(-/-) BMSCs. We found that FGF2 stimulated ATF4 mRNA expression as early as 20 min and increased ATF4 protein expression after three hours of treatment. BMSCs from Fgf2(+/+) and Fgf2(-/-) mice were cultured in osteogenesis medium. We observed reduced alkaline phosphatase staining, decreased mineralized nodules and reduced osteocalcin expression, and reduced expression of ATF4 in Fgf2(-/-) BMSC cultures compared to Fgf2(+/+) BMSCs. This study is the first demonstration that ATF4 expression can be stimulated by FGF2 in osteoblasts and that ATF4 expression is significantly reduced in differentiated Fgf2(-/-) BMSCs. These results suggest that impaired bone mass and bone formation in Fgf2 null mice may be due in part to reduced ATF4 expression.
Collapse
|
42
|
Zhang N, Khachigian LM. Injury-induced platelet-derived growth factor receptor-alpha expression mediated by interleukin-1beta (IL-1beta) release and cooperative transactivation by NF-kappaB and ATF-4: IL-1beta facilitates HDAC-1/2 dissociation from promoter. J Biol Chem 2009; 284:27933-27943. [PMID: 19648113 DOI: 10.1074/jbc.m109.025742] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Platelet-derived growth factors are a family of potent mitogens and chemoattractants for fibroblasts and other cells of mesenchymal origin. Platelet-derived growth factor (PDGF) dimeric ligands (composed of A-, B-, C-, and D-chains) exert their biological activity through high affinity interactions with cell surface receptor subunits (alpha and beta). PDGF-receptor-alpha is widely implicated in the pathogenesis of hyperplastic fibrotic disease, yet the molecular mechanisms controlling its expression in response to injury are poorly understood. Here we show that PDGF-R alpha expression is induced in fibroblasts by mechanical injury and interleukin (IL)-1beta, which was abolished by neutralizing IL-1beta antibodies in the culture supernatant or inhibitors of NF-kappaB. Chromatin immunoprecipitation and electrophoretic mobility shift assays revealed the existence of a new NF-kappaB binding site at -531/-521 bp in the PDGF-R alpha promoter. We have recently shown that ATF-4 is also induced by injury (Malabanan, K. P., Kanellakis, P., Bobik, A., and Khachigian, L. M. (2008) Circ. Res. 103, 378-387), and we demonstrate here that ATF-4 binds a novel element -259/-254 and stimulates PDGF-R alpha transcription. ATF-4 and NF-kappaB interact, occupy the PDGF-R alpha promoter, and induce PDGF-R alpha transcription in a cooperative manner. IL-1beta facilitates the dissociation of histone deacetylase (HDAC)-1/2 from the PDGF-R alpha promoter, whereas the HDAC inhibitors suberoylanilide hydroxamic acid and trichostatin A potentiate IL-1beta induction of PDGF-R alpha transcription. These findings, taken together, demonstrate that injury stimulates IL-1beta secretion by fibroblasts, which activates NF-kappaB and ATF-4 and stimulates interaction with the PDGF-R alpha promoter, triggering PDGF-R alpha transcription. Physical and functional interactions between NF-kappaB and ATF-4 have not been reported in any gene. This is also the first report of HDAC regulation of PDGF-R alpha transcription.
Collapse
Affiliation(s)
- Ning Zhang
- Centre for Vascular Research, University of New South Wales, Sydney, NSW 2052, Australia
| | - Levon M Khachigian
- Centre for Vascular Research, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
43
|
Zhou B, Ma Q, Kong SW, Hu Y, Campbell PH, McGowan FX, Ackerman KG, Wu B, Zhou B, Tevosian SG, Pu WT. Fog2 is critical for cardiac function and maintenance of coronary vasculature in the adult mouse heart. J Clin Invest 2009; 119:1462-76. [PMID: 19411759 DOI: 10.1172/jci38723] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Accepted: 03/11/2009] [Indexed: 12/27/2022] Open
Abstract
Aberrant transcriptional regulation contributes to the pathogenesis of both congenital and adult forms of heart disease. While the transcriptional regulator friend of Gata 2 (FOG2) is known to be essential for heart morphogenesis and coronary development, its tissue-specific function has not been previously investigated. Additionally, little is known about the role of FOG2 in the adult heart. Here we used spatiotemporally regulated inactivation of Fog2 to delineate its function in both the embryonic and adult mouse heart. Early cardiomyocyte- restricted loss of Fog2 recapitulated the cardiac and coronary defects of the Fog2 germline murine knockouts. Later cardiomyocyte-restricted loss of Fog2 (Fog2MC) did not result in defects in cardiac structure or coronary vessel formation. However, Fog2MC adult mice had severely depressed ventricular function and died at 8-14 weeks. Fog2MC adult hearts displayed a paucity of coronary vessels, associated with myocardial hypoxia, increased cardiomyocyte apoptosis, and cardiac fibrosis. Induced inactivation of Fog2 in the adult mouse heart resulted in similar phenotypes, as did ablation of the FOG2 interaction with the transcription factor GATA4. Loss of the FOG2 or FOG2-GATA4 interaction altered the expression of a panel of angiogenesis-related genes. Collectively, our data indicate that FOG2 regulates adult heart function and coronary angiogenesis.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Cardiology, Children's Hospital Boston and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Li J, Wang JJ, Yu Q, Wang M, Zhang SX. Endoplasmic reticulum stress is implicated in retinal inflammation and diabetic retinopathy. FEBS Lett 2009; 583:1521-7. [PMID: 19364508 DOI: 10.1016/j.febslet.2009.04.007] [Citation(s) in RCA: 184] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 03/23/2009] [Accepted: 04/03/2009] [Indexed: 02/06/2023]
Abstract
Diabetic retinopathy is a chronic low-grade inflammatory disease; however, the mechanisms remain elusive. In the present study, we demonstrated that endoplasmic reticulum (ER) stress was activated in the retina in animal models of diabetes and oxygen-induced retinopathy (OIR). Induction of ER stress by tunicamycin resulted in significantly increased expression of inflammatory molecules in the retina. Inhibition of ER stress by chemical chaperone 4-phenyl butyric acid ameliorated inflammation in cultured human retinal endothelial cells exposed to hypoxia, and in the retinas of diabetic and OIR mice. These findings indicate that ER stress is a potential mediator of retinal inflammation in diabetic retinopathy.
Collapse
Affiliation(s)
- Jingming Li
- Harold Hamm Oklahoma Diabetes Center and Section of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center, 941 Stanton L. Young Blvd., BSEB 331A, Oklahoma City, OK 73104, USA
| | | | | | | | | |
Collapse
|
45
|
Abcouwer SF, Roybal CN. Regulation of vascular endothelial growth factor A by activating transcription factor 4. Circ Res 2008; 103:e118; author reply e119. [PMID: 18948622 DOI: 10.1161/circresaha.108.186544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
46
|
Khachigian LM, Bobik A, Kanellakis P, Malabanan K. Response to Abcouwer and Roybal. Circ Res 2008. [DOI: 10.1161/circresaha.108.187559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Levon M. Khachigian
- Centre for Vascular Research, University of New South Wales, Sydney, Australia,
| | | | | | - Kristine Malabanan
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| |
Collapse
|
47
|
|