51
|
Wu JH, Zhou YF, Hong CD, Chen AQ, Luo Y, Mao L, Xia YP, He QW, Jin HJ, Huang M, Li YN, Hu B. Semaphorin-3A protects against neointimal hyperplasia after vascular injury. EBioMedicine 2018; 39:95-108. [PMID: 30579864 PMCID: PMC6355729 DOI: 10.1016/j.ebiom.2018.12.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Neointimal hyperplasia is a prominent pathological event during in-stent restenosis. Phenotype switching of vascular smooth muscle cells (VSMCs) from a differentiated/contractile to a dedifferentiated/synthetic phenotype, accompanied by migration and proliferation of VSMCs play an important role in neointimal hyperplasia. However, the molecular mechanisms underlying phenotype switching of VSMCs have yet to be fully understood. METHODS The mouse carotid artery ligation model was established to evaluate Sema3A expression and its role during neointimal hyperplasia in vivo. Bioinformatics analysis, chromatin immunoprecipitation (ChIP) assays and promoter-luciferase reporter assays were used to examine regulatory mechanism of Sema3A expression. SiRNA transfection and lentivirus infection were performed to regulate Sema3A expression. EdU assays, Wound-healing scratch experiments and Transwell migration assays were used to assess VSMC proliferation and migration. FINDINGS In this study, we found that semaphorin-3A (Sema3A) was significantly downregulated in VSMCs during neointimal hyperplasia after vascular injury in mice and in human atherosclerotic plaques. Meanwhile, Sema3A was transcriptionally downregulated by PDGF-BB via p53 in VSMCs. Furthermore, we found that overexpression of Sema3A inhibited VSMC proliferation and migration, as well as increasing differentiated gene expression. Mechanistically, Sema3A increased the NRP1-plexin-A1 complex and decreased the NRP1-PDGFRβ complex, thus inhibiting phosphorylation of PDGFRβ. Moreover, we found that overexpression of Sema3A suppressed neointimal hyperplasia after vascular injury in vivo. INTERPRETATION These results suggest that local delivery of Sema3A may act as a novel therapeutic option to prevent in-stent restenosis.
Collapse
Affiliation(s)
- Jie-Hong Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Fan Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Can-Dong Hong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - An-Qi Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Luo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Peng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quan-Wei He
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui-Juan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Huang
- Department of Neurology, the People's Hospital of China Three Gorges University, Institute of Translational Neuroscience, Three Gorges University College of Medicine, Yichang 443002, China
| | - Ya-Nan Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
52
|
Hu S, Zhu L. Semaphorins and Their Receptors: From Axonal Guidance to Atherosclerosis. Front Physiol 2018; 9:1236. [PMID: 30405423 PMCID: PMC6196129 DOI: 10.3389/fphys.2018.01236] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 08/15/2018] [Indexed: 12/24/2022] Open
Abstract
Semaphorins are a large family of secreted, transmembrane, or GPI-anchored proteins initially identified as axon guidance cues signaling through their receptors, neuropilins, and plexins. Emerging evidence suggests that beyond the guidance, they also function in a broad spectrum of pathophysiological conditions, including atherosclerosis, a vascular inflammatory disease. Particular semaphorin members have been demonstrated to participate in atherosclerosis via eliciting endothelial dysfunction, leukocyte infiltration, monocyte-macrophage retention, platelet hyperreactivity, and neovascularization. In this review, we focus on the role of those semaphorin family members in the development of atherosclerosis and highlight the mechanistic relevance of semaphorins to atherogenesis.
Collapse
Affiliation(s)
- Shuhong Hu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Li Zhu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
53
|
Wischhusen J, Wilson KE, Delcros JG, Molina-Peña R, Gibert B, Jiang S, Ngo J, Goldschneider D, Mehlen P, Willmann JK, Padilla F. Ultrasound molecular imaging as a non-invasive companion diagnostic for netrin-1 interference therapy in breast cancer. Theranostics 2018; 8:5126-5142. [PMID: 30429890 PMCID: PMC6217066 DOI: 10.7150/thno.27221] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023] Open
Abstract
In ultrasound molecular imaging (USMI), ligand-functionalized microbubbles (MBs) are used to visualize vascular endothelial targets. Netrin-1 is upregulated in 60% of metastatic breast cancers and promotes tumor progression. A novel netrin-1 interference therapy requires the assessment of netrin-1 expression prior to treatment. In this study, we studied netrin-1 as a target for USMI and its potential as a companion diagnostic in breast cancer models. Methods: To verify netrin-1 expression and localization, an in vivo immuno-localization approach was applied, in which anti-netrin-1 antibody was injected into living mice 24 h before tumor collection, and revealed with secondary fluorescent antibody for immunofluorescence analysis. Netrin-1 interactions with the cell surface were studied by flow cytometry. Netrin-1-targeted MBs were prepared using MicroMarker Target-Ready (VisualSonics), and validated in in vitro binding assays in static conditions or in a flow chamber using purified netrin-1 protein or netrin-1-expressing cancer cells. In vivo USMI of netrin-1 was validated in nude mice bearing human netrin-1-positive SKBR7 tumors or weakly netrin-1-expressing MDA-MB-231 tumors using the Vevo 2100 small animal imaging device (VisualSonics). USMI feasibility was further tested in transgenic murine FVB/N Tg(MMTV/PyMT634Mul) (MMTV-PyMT) mammary tumors. Results: Netrin-1 co-localized with endothelial CD31 in netrin-1-positive breast tumors. Netrin-1 binding to the surface of endothelial HUVEC and cancer cells was partially mediated by heparan sulfate proteoglycans. MBs targeted with humanized monoclonal anti-netrin-1 antibody bound to netrin-1-expressing cancer cells in static and dynamic conditions. USMI signal was significantly increased with anti-netrin-1 MBs in human SKBR7 breast tumors and transgenic murine MMTV-PyMT mammary tumors compared to signals recorded with either isotype control MBs or after blocking of netrin-1 with humanized monoclonal anti-netrin-1 antibody. In weakly netrin-1-expressing human tumors and normal mammary glands, no difference in imaging signal was observed with anti-netrin-1- and isotype control MBs. Ex vivo analysis confirmed netrin-1 expression in MMTV-PyMT tumors. Conclusions: These results show that USMI allowed reliable detection of netrin-1 on the endothelium of netrin-1-positive human and murine tumors. Significant differences in USMI signal for netrin-1 reflected the significant differences in netrin-1 mRNA & protein expression observed between different breast tumor models. The imaging approach was non-invasive and safe, and provided the netrin-1 expression status in near real-time. Thus, USMI of netrin-1 has the potential to become a companion diagnostic for the stratification of patients for netrin-1 interference therapy in future clinical trials.
Collapse
|
54
|
Luque MCA, Galuppo MK, Capelli-Peixoto J, Stolf BS. CD100 Effects in Macrophages and Its Roles in Atherosclerosis. Front Cardiovasc Med 2018; 5:136. [PMID: 30324109 PMCID: PMC6173139 DOI: 10.3389/fcvm.2018.00136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/07/2018] [Indexed: 12/31/2022] Open
Abstract
CD100 or Sema4D is a protein from the semaphorin family with important roles in the vascular, nervous and immune systems. It may be found as a membrane bound dimer or as a soluble molecule originated by proteolytic cleavage. Produced by the majority of hematopoietic cells including B and T lymphocytes, natural killer and myeloid cells, as well as endothelial cells, CD100 exerts its actions by binding to different receptors depending on the cell type and on the organism. Cell-to-cell adhesion, angiogenesis, phagocytosis, T cell priming, and antibody production are examples of the many functions of this molecule. Of note, high CD100 serum levels has been found in inflammatory as well as in infectious diseases, but the roles of the protein in the pathogenesis of these diseases has still to be clarified. Macrophages are highly heterogeneous cells present in almost all tissues, which may change their functions in response to microenvironmental conditions. They are key players in the innate and adaptive immune responses and have decisive roles in sterile conditions but also in several diseases such as atherosclerosis, autoimmunity, tumorigenesis, and antitumor responses, among others. Although it is known that macrophages express CD100 and its receptors, few studies have focused on the role of this semaphorin in this cell type or in macrophage-associated diseases. The aim of this review is to critically revise the available data about CD100 and atherosclerosis, with special emphasis on its roles in macrophages and monocytes. We will also describe the few available data on treatments with anti-CD100 antibodies in different diseases. We hope that this review stimulates future studies on the effects of such an important molecule in a cell type with decisive roles in inflammatory diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Maria C A Luque
- Heart Institute, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
55
|
Zhang H, Vreeken D, Bruikman CS, van Zonneveld AJ, van Gils JM. Understanding netrins and semaphorins in mature endothelial cell biology. Pharmacol Res 2018; 137:1-10. [PMID: 30240825 DOI: 10.1016/j.phrs.2018.09.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/15/2018] [Accepted: 09/15/2018] [Indexed: 02/07/2023]
Abstract
Netrins and semaphorins are known as neuronal guidance molecules that are important to the facilitate patterning of the nervous system in embryonic development. In recent years, their function has been broadened to guide development in other systems, including the vascular system, where netrins and semaphorins critically contribute to the development of the vascular system. Evidence is accumulating that these guidance cues are also of critical importance in the biology of the mature endothelium by regulating the maintenance of endothelial quiescence. Here we review our current insights into the roles of netrins and semaphorins in endothelial cell survival, self-renewing, barrier function, response to wall shear stress, and control of the vascular tone. We also provide suggestions for future research into the functions of netrins and semaphorins in mature endothelial cell biology.
Collapse
Affiliation(s)
- Huayu Zhang
- Department of Internal Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Dianne Vreeken
- Department of Internal Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Caroline S Bruikman
- Amsterdam UMC, University of Amsterdam, Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, the Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Janine M van Gils
- Department of Internal Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands.
| |
Collapse
|
56
|
Wu JH, Li Y, Zhou YF, Haslam J, Elvis ON, Mao L, Xia YP, Hu B. Semaphorin-3E attenuates neointimal formation via suppressing VSMCs migration and proliferation. Cardiovasc Res 2018; 113:1763-1775. [PMID: 29016743 DOI: 10.1093/cvr/cvx190] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 09/14/2017] [Indexed: 02/06/2023] Open
Abstract
Aims The migration and proliferation of vascular smooth muscle cells (VSMCs) are crucial events in the neointimal formation, a hallmark of atherosclerosis and restenosis. Semaphorin3E (Sema3E) has been found to be a critical regulator of cell migration and proliferation in many scenarios. However, its role on VSMCs migration and proliferation is unclear. This study aimed to investigate the effect of Sema3E on VSMCs migration, proliferation and neointimal formation, and explore possible mechanisms. Methods and results We found that the expression of Sema3E was progressively decreased during neointimal formation in a carotid ligation model. H&E-staining showed lentivirus-mediated overexpression of Sema3E in carotid ligation area attenuated neointimal formation. Immunofluorescence staining showed that the receptor (PlexinD1) of Sema3E was expressed in vascular walls. In cultured mouse VSMCs, Sema3E inhibited VSMCs migration and proliferation via plexinD1 receptor. The inhibitory effect was mediated, at least in part, by inactivating Rap1-AKT signalling pathways in VSMCs. Moreover, we found that PDGFBB down-regulated the expression of Sema3E in VSMCs and Sema3E notably inhibited the expression of PDGFB in endothelial cells. In addition, the number of Sema3E-positive VSMCs was diminished in plaques of atherosclerotic patients. Results from a public GEO microarray database showed a negative correlation between Sema3E and PDGFB transcriptional levels in the human plaques examined. Conclusion Our study demonstrates that Sema3E/plexinD1 inhibits proliferation and migration of VSMCs via inactivation of Rap1-AKT signalling pathways. The mutual inhibition between PDGF-BB and Sema3E after vascular injury plays a critical role in the process of neointimal formation.
Collapse
Affiliation(s)
- Jie-Hong Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanan Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yi-Fan Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - James Haslam
- Swansea College of Medicine, Swansea University, Singleton Park, Swansea, Wales SA2 8PP, UK
| | - Opoku Nana Elvis
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuan-Peng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
57
|
Liu Y, Chen T, Xing J. Effect of rosuvastatin on the expression of candidate gene GALNT3 in atherosclerosis. Exp Ther Med 2018; 15:4880-4884. [PMID: 29805509 PMCID: PMC5952099 DOI: 10.3892/etm.2018.6008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 02/01/2018] [Indexed: 11/05/2022] Open
Abstract
The effect of rosuvastatin on the expression of candidate gene polypeptide N-acetylgalactosaminyltrans ferase 3 (GALNT3) in atherosclerosis was studied. Sixty Wistar rats were randomly divided into the control (n=20), model (atherosclerosis group, n=20) and administration (rosuvastatin group, n=20) groups. The atherosclerosis model was established via injecting D3.6 million units of vitamin per kilogram of body weight and then the rats were fed with high-fat diet for 6 weeks. The total cholesterol, serum triglyceride and nitric oxide contents were detected using the kits, the morphological changes in thoracic aorta were observed via hematoxylin and eosin (H&E) staining, the mRNA expression of candidate gene GALNT3 was detected via reverse transcription-polymerase chain reaction (RT-PCR), and the protein expression of candidate gene GALNT3 was detected via western blot analysis. Compared with those in the control group, the contents of serum triglyceride and total cholesterol in the model group were significantly increased, and then significantly decreased after drug administration. Morphological observation showed that the surface of thoracic aorta was not smooth with endothelial shedding, and the smooth muscle cells were arranged irregularly and their number was obviously increased. Moreover, RT-PCR and western blot analysis revealed that the mRNA and protein expressions of GALNT3 were significantly increased in the administration group. Rosuvastatin can therefore significantly upregulate the expression of candidate gene GALNT3 in atherosclerosis, thereby reducing the incidence of atherosclerosis.
Collapse
Affiliation(s)
- Yanshao Liu
- Department of Cardiovascular Medicine, Jinan Zhangqiu District Hospital of Traditional Chinese Medicine, Jinan, Shandong 250200, P.R. China
| | - Tao Chen
- Department of Cardiovascular Medicine, Jinan Zhangqiu District Hospital of Traditional Chinese Medicine, Jinan, Shandong 250200, P.R. China
| | - Jieyong Xing
- Department of Cardiovascular Medicine, Jinan Zhangqiu District Hospital of Traditional Chinese Medicine, Jinan, Shandong 250200, P.R. China
| |
Collapse
|
58
|
Jones EA, Lehoux S. Shear stress, arterial identity and atherosclerosis. Thromb Haemost 2018; 115:467-73. [DOI: 10.1160/th15-10-0791] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 12/01/2015] [Indexed: 01/23/2023]
Abstract
SummaryIn the developing embryo, the vasculature first takes the form of a web-like network called the vascular plexus. Arterial and venous differentiation is subsequently guided by the specific expression of genes in the endothelial cells that provide spatial and temporal cues for development. Notch1/4, Notch ligand delta-like 4 (Dll4), and Notch downstream effectors are typically expressed in arterial cells along with EphrinB2, whereas chicken ovalbumin upstream promoter transcription factor II (COUP-TFII) and EphB4 characterise vein endothelial cells. Haemodynamic forces (blood pressure and blood flow) also contribute importantly to vascular remodelling. Early arteriovenous differentiation and local blood flow may hold the key to future inflammatory diseases. Indeed, despite the fact that atherosclerosis risk factors such as smoking, hypertension, hypercholesterolaemia, and diabetes all induce endothelial cell dysfunction throughout the vasculature, plaques develop only in arteries, and they localise essentially in vessel branch points, curvatures and bifurcations, where blood flow (and consequently shear stress) is low or oscillatory. Arterial segments exposed to high blood flow (and high laminar shear stress) tend to remain plaque-free. These observations have led many to investigate what particular properties of arterial or venous endothelial cells confer susceptibility or protection from plaque formation, and how that might interact with a particular shear stress environment.
Collapse
|
59
|
Chistiakov DA, Grechko AV, Myasoedova VA, Melnichenko AA, Orekhov AN. The role of monocytosis and neutrophilia in atherosclerosis. J Cell Mol Med 2018; 22:1366-1382. [PMID: 29364567 PMCID: PMC5824421 DOI: 10.1111/jcmm.13462] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
Monocytosis and neutrophilia are frequent events in atherosclerosis. These phenomena arise from the increased proliferation of hematopoietic stem and multipotential progenitor cells (HSPCs) and HSPC mobilization from the bone marrow to other immune organs and circulation. High cholesterol and inflammatory signals promote HSPC proliferation and preferential differentiation to the myeloid precursors (i.e., myelopoiesis) that than give rise to pro-inflammatory immune cells. These cells accumulate in the plaques thereby enhancing vascular inflammation and contributing to further lesion progression. Studies in animal models of atherosclerosis showed that manipulation with HSPC proliferation and differentiation through the activation of LXR-dependent mechanisms and restoration of cholesterol efflux may have a significant therapeutic potential.
Collapse
MESH Headings
- Animals
- Atherosclerosis/genetics
- Atherosclerosis/immunology
- Atherosclerosis/pathology
- Bone Marrow/immunology
- Bone Marrow/pathology
- Cell Differentiation
- Cell Proliferation
- Cholesterol/immunology
- Disease Models, Animal
- Gene Expression Regulation
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/pathology
- Humans
- Hypercholesterolemia/genetics
- Hypercholesterolemia/immunology
- Hypercholesterolemia/pathology
- Liver X Receptors/genetics
- Liver X Receptors/immunology
- Mice
- Monocytes/immunology
- Monocytes/pathology
- Multipotent Stem Cells/immunology
- Multipotent Stem Cells/pathology
- Neutrophils/immunology
- Neutrophils/pathology
- Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/immunology
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/immunology
- Plaque, Atherosclerotic/pathology
Collapse
Affiliation(s)
- Dimitry A. Chistiakov
- Department of NeurochemistryDivision of Basic and Applied NeurobiologySerbsky Federal Medical Research Center of Psychiatry and NarcologyMoscowRussia
| | - Andrey V. Grechko
- Federal Scientific Clinical Center for Resuscitation and RehabilitationMoscowRussia
| | - Veronika A. Myasoedova
- Skolkovo Innovative CenterInstitute for Atherosclerosis ResearchMoscowRussia
- Laboratory of AngiopathologyInstitute of General Pathology and PathophysiologyRussian Academy of SciencesMoscowRussia
| | - Alexandra A. Melnichenko
- Skolkovo Innovative CenterInstitute for Atherosclerosis ResearchMoscowRussia
- Laboratory of AngiopathologyInstitute of General Pathology and PathophysiologyRussian Academy of SciencesMoscowRussia
| | - Alexander N. Orekhov
- Skolkovo Innovative CenterInstitute for Atherosclerosis ResearchMoscowRussia
- Laboratory of AngiopathologyInstitute of General Pathology and PathophysiologyRussian Academy of SciencesMoscowRussia
| |
Collapse
|
60
|
Hu S, Liu Y, You T, Heath J, Xu L, Zheng X, Wang A, Wang Y, Li F, Yang F, Cao Y, Zhang H, van Gils JM, van Zonneveld AJ, Jo H, Wu Q, Zhang Y, Tang C, Zhu L. Vascular Semaphorin 7A Upregulation by Disturbed Flow Promotes Atherosclerosis Through Endothelial β1 Integrin. Arterioscler Thromb Vasc Biol 2017; 38:335-343. [PMID: 29269512 DOI: 10.1161/atvbaha.117.310491] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/06/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Accumulating evidence suggests a role of semaphorins in vascular homeostasis. Here, we investigate the role of Sema7A (semaphorin 7A) in atherosclerosis and its underlying mechanism. APPROACH AND RESULTS Using genetically engineered Sema7A-/-ApoE-/- mice, we showed that deletion of Sema7A attenuates atherosclerotic plaque formation primarily in the aorta of ApoE-/- mice on a high-fat diet. A higher level of Sema7A in the atheroprone lesser curvature suggests a correlation of Sema7A with disturbed flow. This notion is supported by elevated Sema7A expression in human umbilical venous endothelial cells either subjected to oscillatory shear stress or treated with the PKA (protein kinase A)/CREB (cAMP response element-binding protein) inhibitor H89 (N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide·2HCl hydrate). Further studies using the partial carotid artery ligation model showed that disturbed flow in the left carotid artery of Sema7A+/+ApoE-/- mice promoted the expression of endothelial Sema7A and cell adhesion molecules, leukocyte adhesion, and plaque formation, whereas such changes were attenuated in Sema7A-/-ApoE-/- mice. Further studies showed that blockage of β1 integrin, a known Sema7A receptor, or inhibition of FAK (focal adhesion kinase), MEK1/2 (mitogen-activated protein kinase kinase 1/2), or NF-κB (nuclear factor-κB) significantly reduced the expression of cell adhesion molecules and THP-1 (human acute monocytic leukemia cell line) monocyte adhesion in Sema7A-overexpressing human umbilical venous endothelial cells. Studies using chimeric mice suggest that vascular, most likely endothelial, Sema7A plays a major role in atherogenesis. CONCLUSIONS Our findings indicate a significant role of Sema7A in atherosclerosis by mediating endothelial dysfunction in a β1 integrin-dependent manner.
Collapse
Affiliation(s)
- Shuhong Hu
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Yifei Liu
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Tao You
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Jack Heath
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Linru Xu
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Xiaowei Zheng
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Aili Wang
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Yinyan Wang
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Fengchan Li
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Fei Yang
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Yiren Cao
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Huayu Zhang
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Janine M van Gils
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Anton Jan van Zonneveld
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Hanjoong Jo
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Qingyu Wu
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Yonghong Zhang
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Chaojun Tang
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.).
| | - Li Zhu
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.).
| |
Collapse
|
61
|
Toque HA, Fernandez-Flores A, Mohamed R, Caldwell RB, Ramesh G, Caldwell RW. Netrin-1 is a novel regulator of vascular endothelial function in diabetes. PLoS One 2017; 12:e0186734. [PMID: 29059224 PMCID: PMC5653335 DOI: 10.1371/journal.pone.0186734] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 10/08/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Netrin-1, a secreted laminin-like protein identified as an axon guidance molecule, has been shown to be of critical importance in the cardiovascular system. Recent studies have revealed pro-angiogenic, anti-apoptotic and anti-inflammatory properties of netrin-1 as well as cardioprotective actions against myocardial injury in diabetic mice. AIM To examine the role of netrin-1 in diabetes-and high glucose (HG)-induced vascular endothelial dysfunction (VED) using netrin-1 transgenic mice (Tg3) and cultured bovine aortic endothelial cells (BAEC). MAIN OUTCOME Overexpression of netrin-1 prevented diabetes-induced VED in aorta from diabetic mice and netrin-1 treatment attenuated HG-induced impairment of nitric oxide synthase (NOS) function in BAECs. METHODS AND RESULTS Experiments were performed in Tg3 and littermate control (WT) mice rendered diabetic with streptozotocin (STZ) and in BAECs treated with HG (25 mmol/L). Levels of netrin-1 and its receptor DCC, markers of inflammation and apoptosis and vascular function were assessed in aortas from diabetic and non-diabetic Tg3 and WT mice. Vascular netrin-1 in WT mice was reduced under diabetic conditions. Aortas from non-diabetic Tg3 and WT mice showed similar maximum endothelium-dependent relaxation (MEDR) (83% and 87%, respectively). MEDR was markedly impaired in aorta from diabetic WT mice (51%). This effect was significantly blunted in Tg3 diabetic aortas (70%). Improved vascular relaxation in Tg3 diabetic mice was associated with increased levels of phospho-ERK1/2 and reduced levels of oxidant stress, NFκB, COX-2, p16INK4A, cleaved caspase-3 and p16 and p53 mRNA. Netrin-1 treatment prevented the HG-induced decrease in NO production and elevation of oxidative stress and apoptosis in BAECs. CONCLUSIONS Diabetes decreases aortic levels of netrin-1. However, overexpression of netrin-1 attenuates diabetes-induced VED and limits the reduction of NO levels, while increasing expression of p-ERK1/2, and suppressing oxidative stress and inflammatory and apoptotic processes. Enhancement of netrin-1 function may be a useful therapeutic means for preventing vascular dysfunction in diabetes.
Collapse
Affiliation(s)
- Haroldo A. Toque
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
- * E-mail:
| | - Aracely Fernandez-Flores
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Riyaz Mohamed
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Ruth B. Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
| | - Ganesan Ramesh
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - R. William Caldwell
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| |
Collapse
|
62
|
Layne K, Goodman T, Ferro A, Passacquale G. The effect of aspirin on circulating netrin-1 levels in humans is dependent on the inflammatory status of the vascular endothelium. Oncotarget 2017; 8:86548-86555. [PMID: 29156815 PMCID: PMC5689705 DOI: 10.18632/oncotarget.21240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/07/2017] [Indexed: 01/19/2023] Open
Abstract
In atherosclerotic animal models, the cyclo-oxygenase (COX)-inhibitor aspirin counteracts downregulation of endothelial-derived netrin-1, thus reducing arterial inflammation. We here explored the effect of aspirin on netrin-1 in healthy subjects undergoing influenza immunisation, which is an established experimental model of inflammation-related endothelial dysfunction. Our data showed that netrin-1 undergoes reduction (-29.25% from baseline; p=0.0017) in the presence of endothelial activation (VCAM-1 rose by 9.98% 2-days post-vaccination; p=0.0022). Aspirin counteracted vaccine-induced endothelial activation and reduction of netrin-1 in a dose-dependent manner (-3.06% and -17.03% from baseline at a dose of 300mg and 75mg respectively; p=0.0465 and p>0.05 vs untreated). Clopidogrel, which was used as a comparator due to its similar anti-platelet activity, also reduced endothelial activation but, unlike aspirin, enhanced netrin-1 levels (+20.96% from baseline; p=0.0033 vs untreated). A correlation analysis incorporating cytokines, hs-CRP, VCAM-1, TXB2 and PGE2, showed that changes in netrin-1 were directly related to PGE2 variations only (r=0.6103; p=0.0002). In a separate population of 40 healthy unimmunised volunteers, 28-day treatment with aspirin 300mg reduced netrin-1 (-18.76% from baseline; p=0.0012) without affecting endothelial markers or hs-CRP; as expected, aspirin suppressed TXB2 and PGE2. Netrin-1 and PGE2 levels were directly related (r=0.358; p=0.0015), but other parameters including TXB2, hs-CRP and endothelial markers, were not. In conclusion, aspirin counteracts downregulation of netrin-1 following endothelial dysfunction due to its anti-inflammatory effect on the activated endothelium. However, inhibition of COX-dependent prostanoids negatively modulates netrin-1 synthesis in healthy subjects, and this could give rise to aspirin-dependent reduction in netrin-1 under steady state conditions.
Collapse
Affiliation(s)
- Kerry Layne
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Timothy Goodman
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Albert Ferro
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Gabriella Passacquale
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| |
Collapse
|
63
|
Feinstein J, Ramkhelawon B. Netrins & Semaphorins: Novel regulators of the immune response. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3183-3189. [PMID: 28918114 DOI: 10.1016/j.bbadis.2017.09.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 12/26/2022]
Abstract
Netrins and semaphorins, members of the neuronal guidance cue family, exhibit a rich biology with significant roles that extend beyond chemotactic guidance of the axons to build the neuronal patterns of the body. Screening of adult tissues and specific cellular subsets have illuminated that these proteins are also abundantly expressed under both steady state and pathological scenarios. This observation suggests that, in addition to their role in the development of the axonal tree, these proteins possess additional novel functions in adult physiopathology. Notably, a series of striking evidence has emerged in the literature describing their roles as potent regulators of both innate and adaptive immunity, providing extra dimension to our knowledge of neuronal guidance cues. In this review, we summarize the key complex roles of netrins and semaphorins outside the central nervous system (CNS) with focus on their immunomodulatory functions that impact pathophysiological conditions.
Collapse
Affiliation(s)
- Jordyn Feinstein
- Division of Vascular Surgery, Department of Surgery, New York University School of Medicine, 530 First Avenue, New York, NY 10016, USA; Department of Cell Biology, New York University School of Medicine, 530 First Avenue, New York, NY 10016, USA
| | - Bhama Ramkhelawon
- Division of Vascular Surgery, Department of Surgery, New York University School of Medicine, 530 First Avenue, New York, NY 10016, USA; Department of Cell Biology, New York University School of Medicine, 530 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
64
|
Nguyen MA, Karunakaran D, Geoffrion M, Cheng HS, Tandoc K, Perisic Matic L, Hedin U, Maegdefessel L, Fish JE, Rayner KJ. Extracellular Vesicles Secreted by Atherogenic Macrophages Transfer MicroRNA to Inhibit Cell Migration. Arterioscler Thromb Vasc Biol 2017; 38:49-63. [PMID: 28882869 DOI: 10.1161/atvbaha.117.309795] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 08/18/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVE During inflammation, macrophages secrete vesicles carrying RNA, protein, and lipids as a form of extracellular communication. In the vessel wall, extracellular vesicles (EVs) have been shown to be transferred between vascular cells during atherosclerosis; however, the role of macrophage-derived EVs in atherogenesis is not known. Here, we hypothesize that atherogenic macrophages secrete microRNAs (miRNAs) in EVs to mediate cell-cell communication and promote proinflammatory and proatherogenic phenotypes in recipient cells. APPROACH AND RESULTS We isolated EVs from mouse and human macrophages treated with an atherogenic stimulus (oxidized low-density lipoprotein) and characterized the EV miRNA expression profile. We confirmed the enrichment of miR-146a, miR-128, miR-185, miR-365, and miR-503 in atherogenic EVs compared with controls and demonstrate that these EVs are taken up and transfer exogenous miRNA to naive recipient macrophages. Bioinformatic pathway analysis suggests that atherogenic EV miRNAs are predicted to target genes involved in cell migration and adhesion pathways, and indeed delivery of EVs to naive macrophages reduced macrophage migration both in vitro and in vivo. Inhibition of miR-146a, the most enriched miRNA in atherogenic EVs, reduced the inhibitory effect of EVs on macrophage migratory capacity. EV-mediated delivery of miR-146a repressed the expression of target genes IGF2BP1 (insulin-like growth factor 2 mRNA-binding protein 1) and HuR (human antigen R or ELAV-like RNA-binding protein 1) in recipient cells, and knockdown of IGF2BP1 and HuR using short interfering RNA greatly reduced macrophage migration, highlighting the importance of these EV-miRNA targets in regulating macrophage motility. CONCLUSIONS EV-derived miRNAs from atherogenic macrophages, in particular miR-146a, may accelerate the development of atherosclerosis by decreasing cell migration and promoting macrophage entrapment in the vessel wall.
Collapse
Affiliation(s)
- My-Anh Nguyen
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Denuja Karunakaran
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Michèle Geoffrion
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Henry S Cheng
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Kristofferson Tandoc
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Ljubica Perisic Matic
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Ulf Hedin
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Lars Maegdefessel
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Jason E Fish
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Katey J Rayner
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.).
| |
Collapse
|
65
|
Muñoz JC, Martín R, Alonso C, Gutiérrez B, Nieto ML. Relation between serum levels of chemotaxis-related factors and the presence of coronary artery calcification as expression of subclinical atherosclerosis. Clin Biochem 2017; 50:1048-1055. [PMID: 28830786 DOI: 10.1016/j.clinbiochem.2017.08.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/02/2017] [Accepted: 08/19/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Atherosclerotic plaque formation is characterized by recruitment of monocytes/macrophages, which contributes to its calcification by releasing pro-osteogenic cytokines. Chemotaxis-related proteins, including netrin-1, gremlin-1 and macrophage inflammatory protein-1β (MIP-1β), regulate immune cell migration. However, their relation with the presence of subclinical atherosclerosis, assessed by measures of coronary artery calcifications (CAC) in patients without known coronary artery disease (CAD), remains unclear. AIMS To examine whether these chemoattractant-related proteins are associated with the presence of CAC in patients without known CAD. METHODS A retrospective case-control observational study was conducted in 120 outpatients without CAD, undergoing a CAC evaluation by computed tomography with the Agatston Calcium score, categorized as CAC- (none) and CAC+ (≥1). Serum biomarkers were quantified by ELISA. RESULTS Lpa, dyslipidaemia and smoking were significantly higher (p=0.006, p≤0.0001 and p=0.001, respectively) in CAC+ patients. Serum netrin-1 levels were lower in CAC+ than in CAC- patients (196.8±127.8pg/ml versus 748.3±103.2pg/ml, p≤0.0001), and a similar pattern was found for gremlin-1 (1.14±0.39ng/ml versus 4.33±1.20ng/ml, p≤0.0001). However, TNFα and MIP-1β were strongly upregulated in CAC+ patients (447.56±74pg/ml versus 1104±144pg/ml and 402.00±94pg/ml versus 905.0±101.6pg/ml, respectively, p≤0.001). Multivariate analyses revealed that low netrin-1 and gremlin-1 levels and high TNFα and MIP-1β amounts were associated with CAC presence, after adjustment for clinical and biochemical variables. CONCLUSIONS We found a netrin-1 and gremlin-1 deficiency and a TNFα and MIP-1β overproduction in CAC+ patients' serum. These proteins may be used to identify individuals with subclinical atherosclerosis. Further research is warranted in a larger cohort of patients to establish these chemotactic-related proteins as biomarkers that improve CAD risk stratification.
Collapse
Affiliation(s)
- Juan Carlos Muñoz
- Servicio de Cardiología, Hospital Universitario Rio Hortega, Valladolid, Spain
| | - Rubén Martín
- Instituto de Biología y Genética Molecular, CSIC-UVa, Valladolid, Spain
| | - Carmen Alonso
- Servicio de Radiología, Hospital Universitario Rio Hortega, Valladolid, Spain
| | - Beatriz Gutiérrez
- Instituto de Biología y Genética Molecular, CSIC-UVa, Valladolid, Spain
| | - María Luisa Nieto
- Instituto de Biología y Genética Molecular, CSIC-UVa, Valladolid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV). Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
66
|
Maruyama K, Takemura N, Martino MM, Kondo T, Akira S. Netrins as prophylactic targets in skeletal diseases: A double-edged sword? Pharmacol Res 2017; 122:46-52. [PMID: 28576474 DOI: 10.1016/j.phrs.2017.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/11/2017] [Accepted: 05/11/2017] [Indexed: 12/31/2022]
Abstract
The netrin family of proteins are involved in axon guidance during central nervous system development. In vertebrates, two membrane bound forms and five secreted forms of netrin have been reported. In addition to their critical role in neural morphogenesis, a growing number of reports suggest that netrin family proteins also play a role in inflammatory conditions, angiogenesis, and tumorigenesis. In these processes, Unc5 and DCC family proteins serve as receptors of netrin proteins. Recently, it was reported that some netrin family proteins may be involved in the pathogenesis of skeletal diseases including osteoporosis and arthritis. For example, administration of secreted netrin family proteins such as netrin 1 and netrin 4 has prophylactic potential in pathogenic bone degradation in mice. However, netrin 1 blocking antibody also protects mice from inflammatory bone destruction. Therefore, netrin family proteins are involved in the regulation of bone homeostasis, but their bona fide roles in the skeletal system remain controversial. In this review, we discuss the osteo-innate-immune functions of the netrin family of proteins, and summarize their therapeutic potential.
Collapse
Affiliation(s)
- Kenta Maruyama
- Laboratory of Host Defense Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan, Japan; WPI Immunology Frontier Research Center (IFReC), 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Naoki Takemura
- Department of Mucosal Immunology, School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan; Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Mikaël M Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria, 3800, Australia
| | - Takeshi Kondo
- Laboratory of Host Defense Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan, Japan; WPI Immunology Frontier Research Center (IFReC), 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shizuo Akira
- Laboratory of Host Defense Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan, Japan; WPI Immunology Frontier Research Center (IFReC), 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
67
|
Mirakaj V, Rosenberger P. Immunomodulatory Functions of Neuronal Guidance Proteins. Trends Immunol 2017; 38:444-456. [PMID: 28438491 DOI: 10.1016/j.it.2017.03.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023]
Abstract
Neuronal guidance proteins (NGPs) were originally identified for their role during the embryonic development of the nervous system. Recent years have seen the discovery of NGP functions during immune responses. In this context, NGPs were demonstrated to control leukocyte migration and the release of cytokines during conditions of acute inflammation, such as lung injury or sepsis. However, NGPs also display potent actions in the resolution of inflammation, chronic inflammatory conditions, the development of atherosclerosis, and during ischemia followed by reperfusion. Here, we provide an overview of the current state of knowledge about the role of NGPs in the immune system and describe their immunomodulatory function.
Collapse
Affiliation(s)
- Valbona Mirakaj
- Department of Anesthesia and Intensive Care Medicine, Tübingen University Hospital, Faculty of Medicine, Eberhard-Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany.
| | - Peter Rosenberger
- Department of Anesthesia and Intensive Care Medicine, Tübingen University Hospital, Faculty of Medicine, Eberhard-Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany.
| |
Collapse
|
68
|
Integrin signaling in atherosclerosis. Cell Mol Life Sci 2017; 74:2263-2282. [PMID: 28246700 DOI: 10.1007/s00018-017-2490-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/24/2017] [Accepted: 02/15/2017] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic lipid-driven inflammatory disease affecting large arteries, represents the primary cause of cardiovascular disease in the world. The local remodeling of the vessel intima during atherosclerosis involves the modulation of vascular cell phenotype, alteration of cell migration and proliferation, and propagation of local extracellular matrix remodeling. All of these responses represent targets of the integrin family of cell adhesion receptors. As such, alterations in integrin signaling affect multiple aspects of atherosclerosis, from the earliest induction of inflammation to the development of advanced fibrotic plaques. Integrin signaling has been shown to regulate endothelial phenotype, facilitate leukocyte homing, affect leukocyte function, and drive smooth muscle fibroproliferative remodeling. In addition, integrin signaling in platelets contributes to the thrombotic complications that typically drive the clinical manifestation of cardiovascular disease. In this review, we examine the current literature on integrin regulation of atherosclerotic plaque development and the suitability of integrins as potential therapeutic targets to limit cardiovascular disease and its complications.
Collapse
|
69
|
Wong WT, Ma S, Tian XY, Gonzalez AB, Ebong EE, Shen H. Targeted Delivery of Shear Stress-Inducible Micrornas by Nanoparticles to Prevent Vulnerable Atherosclerotic Lesions. Methodist Debakey Cardiovasc J 2016; 12:152-156. [PMID: 27826369 DOI: 10.14797/mdcj-12-3-152] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular wall disease, and endothelial cell dysfunction plays an important role in its development and progression. Under the influence of laminar shear stress, however, the endothelium releases homeostatic factors such as nitric oxide and expresses of vasoprotective microRNAs that are resistant to atherosclerosis. Adhesion molecules such as E-selectin, exhibited on the endothelial surface, recruit monocytes that enter the vessel wall to form foam cells. Accumulation of these foam cells form fatty streaks that may progress to atherosclerotic plaques in the blood vessel wall. Interestingly, E-selectin may also serve as an affinity moiety for targeted drug delivery against atherosclerosis. We have recently developed an E-selectin-targeted platform that enriches therapeutic microRNAs in the inflamed endothelium to inhibit formation of vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Wing Tak Wong
- Houston Methodist Research Institute, Houston, Texas
| | - Shuangtao Ma
- Houston Methodist Research Institute, Houston, Texas
| | - Xiao Yu Tian
- Houston Methodist Research Institute, Houston, Texas
| | | | - Eno E Ebong
- Northeastern University, Boston, Massachusetts
| | - Haifa Shen
- Houston Methodist Research Institute, Houston, Texas
| |
Collapse
|
70
|
Taylor L, Brodermann MH, McCaffary D, Iqbal AJ, Greaves DR. Netrin-1 Reduces Monocyte and Macrophage Chemotaxis towards the Complement Component C5a. PLoS One 2016; 11:e0160685. [PMID: 27509208 PMCID: PMC4980032 DOI: 10.1371/journal.pone.0160685] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/24/2016] [Indexed: 01/24/2023] Open
Abstract
Netrin-1, acting at its cognate receptor UNC5b, has been previously demonstrated to inhibit CC chemokine-induced immune cell migration. In line with this, we found that netrin-1 was able to inhibit CCL2-induced migration of bone marrow derived macrophages (BMDMs). However, whether netrin-1 is capable of inhibiting chemotaxis to a broader range of chemoattractants remains largely unexplored. As our initial experiments demonstrated that RAW264.7 and BMDMs expressed high levels of C5a receptor 1 (C5aR1) on their surface, we aimed to determine the effect of netrin-1 exposure on monocyte/macrophage cell migration induced by C5a, a complement peptide that plays a major role in multiple inflammatory pathologies. Treatment of RAW264.7 macrophages, BMDMs and human monocytes with netrin-1 inhibited their chemotaxis towards C5a, as measured using two different real-time methods. This inhibitory effect was found to be dependent on netrin-1 receptor signalling, as an UNC5b blocking antibody was able to reverse netrin-1 inhibition of C5a induced BMDM migration. Treatment of BMDMs with netrin-1 had no effect on C5aR1 proximal signalling events, as surface C5aR1 expression, internalisation and intracellular Ca2+ release following C5aR1 ligation remained unaffected after netrin-1 exposure. We next examined receptor distal events that occur following C5aR1 activation, but found that netrin-1 was unable to inhibit C5a induced phosphorylation of ERK1/2, Akt and p38, pathways important for cellular migration. Furthermore, netrin-1 treatment had no effect on BMDM cytoskeletal rearrangement following C5a stimulation as determined by microscopy and real-time electrical impedance sensing. Taken together these data highlight that netrin-1 inhibits monocyte and macrophage cell migration, but that the mechanism behind this effect remains unresolved. Nevertheless, netrin-1 and its cognate receptors warrant further investigation as they may represent a potential avenue for the development of novel anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- Lewis Taylor
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | | | - David McCaffary
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Asif Jilani Iqbal
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
71
|
Mediero A, Wilder T, Ramkhelawon B, Moore KJ, Cronstein BN. Netrin-1 and its receptor Unc5b are novel targets for the treatment of inflammatory arthritis. FASEB J 2016; 30:3835-3844. [PMID: 27502509 DOI: 10.1096/fj.201600615r] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/27/2016] [Indexed: 12/20/2022]
Abstract
Rheumatoid arthritis is an autoimmune disease that is characterized by chronic inflammation and destruction of joints. Netrin-1, a chemorepulsant, laminin-like matrix protein, promotes inflammation by preventing macrophage egress from inflamed sites and is required for osteoclast differentiation. We asked whether blockade of Netrin-1 or its receptors [Unc5b and DCC (deleted in colorectal carcinoma)] may be useful therapeutic targets for treatment of inflammatory arthritis. Arthritis was induced in 8-wk-old C57Bl/6 mice by intraperitoneal injection of K/BxN serum. Murine monoclonal antibodies against Netrin-1, Unc5b, or DCC (10 µg/mouse) were injected weekly for 4 wk (n = 10). Paw swelling and thickness were assessed and following euthanasia 2-4 wk after serum transfer, paws were prepared for micro-computed tomography and histology. Paw inflammation was maximal 2 wk after injection. Anti-Netrin-1 or anti-Unc5b, but not anti-DCC, antibodies significantly reduced paw inflammation (clinical score: 9.8 ± 0.8, 10.4 ± 0.9, and 13.5 ± 0.5, respectively vs 16 ± 0 for control; P < 0.001). Micro-computed tomography showed bony erosions in untreated or anti-DCC-treated mice, whereas there were no erosions in anti-Netrin-1/anti-Unc5b-treated-animals. Tartrate-resistant acid phosphatase staining demonstrated a marked decrease in osteoclasts in anti-Netrin-1/anti-Unc5b-treated animals. Immunofluorescence staining revealed a decrease in cathepsin K+ and CD68+ cells in anti-Netrin-1/anti-Unc5b-treated animals. Blockade of Netrin-1/Unc5b by monoclonal antibodies prevents bone destruction and reduces the severity of K/BxN serum transfer-induced arthritis. Netrin-1 may be a novel therapeutic target for treatment of inflammatory bone destruction.-Mediero, A., Wilder, T., Ramkhelawon, B., Moore, K. J., Cronstein, B. N. Netrin-1 and its receptor Unc5b are novel targets for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Aránzazu Mediero
- Bone and Joint Research Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Division of Translational Medicine, Department of Medicine, New York University School of Medicine, New York, New York, USA; and
| | - Tuere Wilder
- Division of Translational Medicine, Department of Medicine, New York University School of Medicine, New York, New York, USA; and
| | - Bhama Ramkhelawon
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - Kathryn J Moore
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - Bruce N Cronstein
- Division of Translational Medicine, Department of Medicine, New York University School of Medicine, New York, New York, USA; and
| |
Collapse
|
72
|
Bradfield PF, Menon A, Miljkovic-Licina M, Lee BP, Fischer N, Fish RJ, Kwak B, Fisher EA, Imhof BA. Divergent JAM-C Expression Accelerates Monocyte-Derived Cell Exit from Atherosclerotic Plaques. PLoS One 2016; 11:e0159679. [PMID: 27442505 PMCID: PMC4956249 DOI: 10.1371/journal.pone.0159679] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis, caused in part by monocytes in plaques, continues to be a disease that afflicts the modern world. Whilst significant steps have been made in treating this chronic inflammatory disease, questions remain on how to prevent monocyte and macrophage accumulation in atherosclerotic plaques. Junctional Adhesion Molecule C (JAM-C) expressed by vascular endothelium directs monocyte transendothelial migration in a unidirectional manner leading to increased inflammation. Here we show that interfering with JAM-C allows reverse-transendothelial migration of monocyte-derived cells, opening the way back out of the inflamed environment. To study the role of JAM-C in plaque regression we used a mouse model of atherosclerosis, and tested the impact of vascular JAM-C expression levels on monocyte reverse transendothelial migration using human cells. Studies in-vitro under inflammatory conditions revealed that overexpression or gene silencing of JAM-C in human endothelium exposed to flow resulted in higher rates of monocyte reverse-transendothelial migration, similar to antibody blockade. We then transplanted atherosclerotic, plaque-containing aortic arches from hyperlipidemic ApoE-/- mice into wild-type normolipidemic recipient mice. JAM-C blockade in the recipients induced greater emigration of monocyte-derived cells and further diminished the size of atherosclerotic plaques. Our findings have shown that JAM-C forms a one-way vascular barrier for leukocyte transendothelial migration only when present at homeostatic copy numbers. We have also shown that blocking JAM-C can reduce the number of atherogenic monocytes/macrophages in plaques by emigration, providing a novel therapeutic strategy for chronic inflammatory pathologies.
Collapse
Affiliation(s)
- Paul F. Bradfield
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
- * E-mail:
| | - Arjun Menon
- Division of Cardiology, New York University Langone Medical Center, New York, New York 10016, United States of America
| | - Marijana Miljkovic-Licina
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
| | - Boris P. Lee
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
| | - Nicolas Fischer
- NovImmune S.A., 14 chemin des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Richard J. Fish
- Department of Genetic Medicine and Development, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva, Switzerland
| | - Brenda Kwak
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
| | - Edward A. Fisher
- Division of Cardiology, New York University Langone Medical Center, New York, New York 10016, United States of America
| | - Beat A. Imhof
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
| |
Collapse
|
73
|
Abstract
Genetic factors contribute importantly to the risk of coronary artery disease (CAD), and in the past decade, there has been major progress in this area. The tools applied include genome-wide association studies encompassing >200,000 individuals complemented by bioinformatic approaches, including 1000 Genomes imputation, expression quantitative trait locus analyses, and interrogation of Encyclopedia of DNA Elements, Roadmap, and other data sets. close to 60 common SNPs (minor allele frequency>0.05) associated with CAD risk and reaching genome-wide significance (P<5 × 10(-8)) have been identified. Furthermore, a total of 202 independent signals in 109 loci have achieved a false discovery rate (q<0.05) and together explain 28% of the estimated heritability of CAD. These data have been used successfully to create genetic risk scores that can improve risk prediction beyond conventional risk factors and identify those individuals who will benefit most from statin therapy. Such information also has important applications in clinical medicine and drug discovery by using a Mendelian randomization approach to interrogate the causal nature of many factors found to associate with CAD risk in epidemiological studies. In contrast to genome-wide association studies, whole-exome sequencing has provided valuable information directly relevant to genes with known roles in plasma lipoprotein metabolism but has, thus far, failed to identify other rare coding variants linked to CAD. Overall, recent studies have led to a broader understanding of the genetic architecture of CAD and demonstrate that it largely derives from the cumulative effect of multiple common risk alleles individually of small effect size rather than rare variants with large effects on CAD risk. Despite this success, there has been limited progress in understanding the function of the novel loci; the majority of which are in noncoding regions of the genome.
Collapse
Affiliation(s)
- Ruth McPherson
- From the Department of Medicine, Atherogenomics Laboratory, Division of Cardiology, Ruddy Canadian Cardiovascular Genetics Center, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.M.); and Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.T.-H.).
| | - Anne Tybjaerg-Hansen
- From the Department of Medicine, Atherogenomics Laboratory, Division of Cardiology, Ruddy Canadian Cardiovascular Genetics Center, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.M.); and Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.T.-H.)
| |
Collapse
|
74
|
Affiliation(s)
- Hong Lu
- From the Saha Cardiovascular Research Center, University of Kentucky, Lexington.
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center, University of Kentucky, Lexington
| |
Collapse
|
75
|
Hordijk PL. Recent insights into endothelial control of leukocyte extravasation. Cell Mol Life Sci 2016; 73:1591-608. [PMID: 26794844 PMCID: PMC11108429 DOI: 10.1007/s00018-016-2136-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 12/30/2022]
Abstract
In the process of leukocyte migration from the circulation across the vascular wall, the crosstalk with endothelial cells that line the blood vessels is essential. It is now firmly established that in endothelial cells important signaling events are initiated upon leukocyte adhesion that impinge on the regulation of cell-cell contact and control the efficiency of transendothelial migration. In addition, several external factors such as shear force and vascular stiffness were recently identified as important regulators of endothelial signaling and, consequently, leukocyte transmigration. Here, I review recent insights into endothelial signaling events that are linked to leukocyte migration across the vessel wall. In this field, protein phosphorylation and Rho-mediated cytoskeletal dynamics are still widely studied using increasingly sophisticated mouse models. In addition, activation of tyrosine phosphatases, changes in endothelial cell stiffness as well as different vascular beds have all been established as important factors in endothelial signaling and leukocyte transmigration. Finally, I address less-well-studied but interesting components in the endothelium that also control transendothelial migration, such as the ephrins and their Eph receptors, that provide novel insights in the complexity associated with this process.
Collapse
Affiliation(s)
- Peter L Hordijk
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Swammerdam Institute for Life Sciences, University of Amsterdam, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands.
- Department of Physiology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
76
|
Allen S, Liu YG, Scott E. Engineering nanomaterials to address cell-mediated inflammation in atherosclerosis. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2016; 2:37-50. [PMID: 27135051 DOI: 10.1007/s40883-016-0012-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is an inflammatory disorder with a pathophysiology driven by both innate and adaptive immunity and a primary cause of cardiovascular disease (CVD) worldwide. Vascular inflammation and accumulation of foam cells and their products induce maturation of atheromas, or plaques, which can rupture by metalloprotease action, leading to ischemic stroke or myocardial infarction. Diverse immune cell populations participate in all stages of plaque maturation, many of which directly influence plaque stability and rupture via inflammatory mechanisms. Current clinical treatments for atherosclerosis focus on lowering serum levels of low-density lipoprotein (LDL) using therapeutics such as statins, administration of antithrombotic drugs, and surgical intervention. Strategies that address cell-mediated inflammation are lacking, and consequently have recently become an area of considerable research focus. Nanomaterials have emerged as highly advantageous tools for these studies, as they can be engineered to target specific inflammatory cell populations, deliver therapeutics of wide-ranging solubilities and enhance analytical methods that include imaging and proteomics. Furthermore, the highly phagocytic nature of antigen presenting cells (APCs), a diverse cell population central to the initiation of immune responses and inflammation, make them particularly amenable to targeting and modulation by nanoscale particulates. Nanomaterials have therefore become essential components of vaccine formulations and treatments for inflammation-driven pathologies like autoimmunity, and present novel opportunities for immunotherapeutic treatments of CVD. Here, we review recent progress in the design and use of nanomaterials for therapeutic assessment and treatment of atherosclerosis. We will focus on promising new approaches that utilize nanomaterials for cell-specific imaging, gene therapy and immunomodulation.
Collapse
Affiliation(s)
- Sean Allen
- Department of Biomedical Engineering, Northwestern University, Evanston IL, USA
| | - Yu-Gang Liu
- Department of Biomedical Engineering, Northwestern University, Evanston IL, USA
| | - Evan Scott
- Department of Biomedical Engineering, Northwestern University, Evanston IL, USA
| |
Collapse
|
77
|
Abstract
Atherosclerosis is a complex chronic disease. The accumulation of myeloid cells in the arterial intima, including macrophages and dendritic cells (DCs), is a feature of early stages of disease. For decades, it has been known that monocyte recruitment to the intima contributes to the burden of lesion macrophages. Yet, this paradigm may require reevaluation in light of recent advances in understanding of tissue macrophage ontogeny, their capacity for self-renewal, as well as observations that macrophages proliferate throughout atherogenesis and that self-renewal is critical for maintenance of macrophages in advanced lesions. The rate of atherosclerotic lesion formation is profoundly influenced by innate and adaptive immunity, which can be regulated locally within atherosclerotic lesions, as well as in secondary lymphoid organs, the bone marrow and the blood. DCs are important modulators of immunity. Advances in the past decade have cemented our understanding of DC subsets, functions, hematopoietic origin, gene expression patterns, transcription factors critical for differentiation, and provided new tools for study of DC biology. The functions of macrophages and DCs overlap to some extent, thus it is important to reassess the contributions of each of these myeloid cells taking into account strict criteria of cell identification, ontogeny, and determine whether their key roles are within atherosclerotic lesions or secondary lymphoid organs. This review will highlight key aspect of macrophage and DC biology, summarize how these cells participate in different stages of atherogenesis and comment on complexities, controversies, and gaps in knowledge in the field.
Collapse
Affiliation(s)
- Myron I. Cybulsky
- From the Division of Advanced Diagnostics, Toronto General Research Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada (M.I.C., C.S.R.); Departments of Laboratory Medicine and Pathobiology (M.I.C., C.S.R.) and Immunology (C.S.R.), University of Toronto, Toronto, Ontario, Canada; and Laboratory of Cellular Physiology and Immunology, Institut de Researches Cliniques de Montréal, Montréal, Québec, Canada (C.C.)
| | - Cheolho Cheong
- From the Division of Advanced Diagnostics, Toronto General Research Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada (M.I.C., C.S.R.); Departments of Laboratory Medicine and Pathobiology (M.I.C., C.S.R.) and Immunology (C.S.R.), University of Toronto, Toronto, Ontario, Canada; and Laboratory of Cellular Physiology and Immunology, Institut de Researches Cliniques de Montréal, Montréal, Québec, Canada (C.C.)
| | - Clinton S. Robbins
- From the Division of Advanced Diagnostics, Toronto General Research Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada (M.I.C., C.S.R.); Departments of Laboratory Medicine and Pathobiology (M.I.C., C.S.R.) and Immunology (C.S.R.), University of Toronto, Toronto, Ontario, Canada; and Laboratory of Cellular Physiology and Immunology, Institut de Researches Cliniques de Montréal, Montréal, Québec, Canada (C.C.)
| |
Collapse
|
78
|
Yin P, Lv H, Zhang L, Zhang L, Tang P. Semaphorin 3A: A Potential Target for Low Back Pain. Front Aging Neurosci 2015; 7:216. [PMID: 26635602 PMCID: PMC4659908 DOI: 10.3389/fnagi.2015.00216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 11/09/2015] [Indexed: 01/08/2023] Open
Abstract
Low back pain is a common disorder. Pathological innervation and intervertebral disc degeneration are two major factors associated with this disease. Semaphorin 3A, originally known for its potent inhibiting effect on axonal outgrowth, is recently found to correlate with disease activity and histological features in some skeletal disorders. Based on its effects on innervation and vascularization, as well as enzyme secretion, we presume that semaphorin 3A may act as a potential target for low back pain.
Collapse
Affiliation(s)
- Pengbin Yin
- Department of Orthopedics, Chinese PLA General Hospital Beijing, China
| | - Houchen Lv
- Department of Orthopedics, Chinese PLA General Hospital Beijing, China
| | - Lihai Zhang
- Department of Orthopedics, Chinese PLA General Hospital Beijing, China
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital Beijing, China
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital Beijing, China
| |
Collapse
|
79
|
Poitz DM, Ende G, Stütz B, Augstein A, Friedrichs J, Brunssen C, Werner C, Strasser RH, Jellinghaus S. EphrinB2/EphA4-mediated activation of endothelial cells increases monocyte adhesion. Mol Immunol 2015; 68:648-56. [PMID: 26552760 DOI: 10.1016/j.molimm.2015.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/09/2015] [Accepted: 10/15/2015] [Indexed: 11/15/2022]
Abstract
The membrane anchored ligand ephrinB2 belongs to the broad Eph/ephrin system and is able to activate different Eph receptors. The Eph receptors belong to the huge group of receptor-tyrosine kinases. Eph receptors as well as their corresponding ephrin ligands are cell-membrane attached proteins. Therefore, direct cell-cell contact is essentially for interaction. It is known that ephrinB2 plays a pivotal role in developmental and in tumour angiogenesis. Previous studies point to a crucial role of the EphA4-receptor in the process of monocyte adhesion. Since ephrinB2 is known as an interaction partner of EphA4, the aim of the present study was to investigate a possible interplay of EphA4-receptor with ephrinB2 during monocyte adhesion to the endothelium. As verified by bulk adhesion assays and atomic-force microscopy based single-cell force spectroscopy, temporary stimulation of endothelial cells from different sources with the soluble ligand ephrinB2 increased monocyte adhesion to endothelial cells. The proadhesive effect of ephrinB2 was independent of an active transcription, but is mediated via the Rho signaling pathway with subsequent modulation of the actin cytoskeleton. Furthermore, ephrinB2 mediated its impact on monocyte adhesion via the receptor EphA4 as shown by siRNA-mediated silencing. Interestingly, ephrinB2 was induced by TNF-α treatment. Silencing of ephrinB2 led to a lowering of the TNF-α mediated monocyte adhesion to endothelial cells. Furthermore, immunohistochemical staining of human atherosclerotic plaque revealed expression of ephrinB2 in macrophages. The results of the present study point to a crucial role of ephrinB2 induced EphA4 forward signaling in the context of monocyte adhesion to endothelial cells. This transcription-independent effect is mediated by Rho signaling induced actin-filament polymerization.
Collapse
Affiliation(s)
- David M Poitz
- Department of Internal Medicine and Cardiology, TU Dresden, Germany.
| | - Georg Ende
- Department of Internal Medicine and Cardiology, TU Dresden, Germany
| | - Beryl Stütz
- Department of Internal Medicine and Cardiology, TU Dresden, Germany
| | - Antje Augstein
- Department of Internal Medicine and Cardiology, TU Dresden, Germany
| | - Jens Friedrichs
- Institute for Biofunctional Polymer Materials Dresden, Leibniz Institute of Polymer Research, Germany
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, TU Dresden, Germany
| | - Carsten Werner
- Institute for Biofunctional Polymer Materials Dresden, Leibniz Institute of Polymer Research, Germany
| | - Ruth H Strasser
- Department of Internal Medicine and Cardiology, TU Dresden, Germany
| | | |
Collapse
|
80
|
Rhodes CJ, Im H, Cao A, Hennigs JK, Wang L, Sa S, Chen PI, Nickel NP, Miyagawa K, Hopper RK, Tojais NF, Li CG, Gu M, Spiekerkoetter E, Xian Z, Chen R, Zhao M, Kaschwich M, Del Rosario PA, Bernstein D, Zamanian RT, Wu JC, Snyder MP, Rabinovitch M. RNA Sequencing Analysis Detection of a Novel Pathway of Endothelial Dysfunction in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2015; 192:356-66. [PMID: 26030479 DOI: 10.1164/rccm.201408-1528oc] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Pulmonary arterial hypertension is characterized by endothelial dysregulation, but global changes in gene expression have not been related to perturbations in function. OBJECTIVES RNA sequencing was used to discriminate changes in transcriptomes of endothelial cells cultured from lungs of patients with idiopathic pulmonary arterial hypertension versus control subjects and to assess the functional significance of major differentially expressed transcripts. METHODS The endothelial transcriptomes from the lungs of seven control subjects and six patients with idiopathic pulmonary arterial hypertension were analyzed. Differentially expressed genes were related to bone morphogenetic protein type 2 receptor (BMPR2) signaling. Those down-regulated were assessed for function in cultured cells and in a transgenic mouse. MEASUREMENTS AND MAIN RESULTS Fold differences in 10 genes were significant (P < 0.05), four increased and six decreased in patients versus control subjects. No patient was mutant for BMPR2. However, knockdown of BMPR2 by siRNA in control pulmonary arterial endothelial cells recapitulated 6 of 10 patient-related gene changes, including decreased collagen IV (COL4A1, COL4A2) and ephrinA1 (EFNA1). Reduction of BMPR2-regulated transcripts was related to decreased β-catenin. Reducing COL4A1, COL4A2, and EFNA1 by siRNA inhibited pulmonary endothelial adhesion, migration, and tube formation. In mice null for the EFNA1 receptor, EphA2, versus control animals, vascular endothelial growth factor receptor blockade and hypoxia caused more severe pulmonary hypertension, judged by elevated right ventricular systolic pressure, right ventricular hypertrophy, and loss of small arteries. CONCLUSIONS The novel relationship between BMPR2 dysfunction and reduced expression of endothelial COL4 and EFNA1 may underlie vulnerability to injury in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Christopher J Rhodes
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Hogune Im
- 2 Cardiovascular Institute.,4 Department of Genetics, and
| | - Aiqin Cao
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Jan K Hennigs
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Lingli Wang
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Silin Sa
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Pin-I Chen
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Nils P Nickel
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Kazuya Miyagawa
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Rachel K Hopper
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Nancy F Tojais
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Caiyun G Li
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Mingxia Gu
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Edda Spiekerkoetter
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,5 Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Zhaoying Xian
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Rui Chen
- 2 Cardiovascular Institute.,4 Department of Genetics, and
| | - Mingming Zhao
- 2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Mark Kaschwich
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Patricia A Del Rosario
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,5 Department of Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Roham T Zamanian
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,5 Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Joseph C Wu
- 2 Cardiovascular Institute.,5 Department of Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Marlene Rabinovitch
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| |
Collapse
|
81
|
Mukovozov I, Huang YW, Zhang Q, Liu GY, Siu A, Sokolskyy Y, Patel S, Hyduk SJ, Kutryk MJB, Cybulsky MI, Robinson LA. The Neurorepellent Slit2 Inhibits Postadhesion Stabilization of Monocytes Tethered to Vascular Endothelial Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:3334-44. [PMID: 26297762 DOI: 10.4049/jimmunol.1500640] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 07/22/2015] [Indexed: 01/22/2023]
Abstract
The secreted neurorepellent Slit2, acting through its transmembrane receptor, Roundabout (Robo)-1, inhibits chemotaxis of varied cell types, including leukocytes, endothelial cells, and vascular smooth muscle cells, toward diverse attractants. The role of Slit2 in regulating the steps involved in recruitment of monocytes in vascular inflammation is not well understood. In this study, we showed that Slit2 inhibited adhesion of monocytic cells to activated human endothelial cells, as well as to immobilized ICAM-1 and VCAM-1. Microfluidic live cell imaging showed that Slit2 inhibited the ability of monocytes tethered to endothelial cells to stabilize their actin-associated anchors and to resist detachment in response to increasing shear forces. Transfection of constitutively active plasmids revealed that Slit2 inhibited postadhesion stabilization of monocytes on endothelial cells by preventing activation of Rac1. We further found that Slit2 inhibited chemotaxis of monocytes toward CXCL12 and CCL2. To determine whether Slit2 and Robo-1 modulate pathologic monocyte recruitment associated with vascular inflammation and cardiovascular disease, we tested PBMC from patients with coronary artery disease. PBMC from these patients had reduced surface levels of Robo-1 compared with healthy age- and sex-matched subjects, and Slit2 failed to inhibit chemotaxis of PBMC of affected patients, but not healthy control subjects, toward CCL2. Furthermore, administration of Slit2 to atherosclerosis-prone LDL receptor-deficient mice inhibited monocyte recruitment to nascent atherosclerotic lesions. These results demonstrate that Slit2 inhibits chemotaxis of monocytes, as well as their ability to stabilize adhesions and resist detachment forces. Slit2 may represent a powerful new tool to inhibit pathologic monocyte recruitment in vascular inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Ilya Mukovozov
- Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Yi-Wei Huang
- Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8, Canada
| | - Qiuwang Zhang
- Division of Cardiology, Keenan Research Center for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada; and
| | - Guang Ying Liu
- Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8, Canada
| | - Allan Siu
- Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Yaroslav Sokolskyy
- Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8, Canada
| | - Sajedabanu Patel
- Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8, Canada
| | - Sharon J Hyduk
- Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Michael J B Kutryk
- Division of Cardiology, Keenan Research Center for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada; and
| | - Myron I Cybulsky
- Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Lisa A Robinson
- Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada;
| |
Collapse
|
82
|
Layne K, Ferro A, Passacquale G. Netrin-1 as a novel therapeutic target in cardiovascular disease: to activate or inhibit? Cardiovasc Res 2015. [PMID: 26209250 DOI: 10.1093/cvr/cvv201] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Netrins are a family of laminin-like proteins, which were initially identified for their role in embryonic axonal guidance. Over recent years, it has become apparent that netrin-1 may additionally be involved in the underlying pathology of several multisystem diseases, making it an attractive potential therapeutic target. It is involved in postnatal angiogenesis, particularly in the context of an ischaemic insult, although there are conflicting reports as to whether netrin-1 acts in a pro- or anti-angiogenic capacity. In atherosclerosis, opposing effects have similarly been reported on plaque progression, due to the ability of netrin-1 to inhibit both macrophage egress from and monocyte ingress into lesions. Netrin-1 has also been shown to exert a cardioprotective action in the context of ischaemia-reperfusion injury following myocardial infarction. Moreover, urinary netrin-1 levels rise in response to acute kidney injury and at a faster rate than traditional markers of renal impairment, highlighting a potential clinical role for netrin-1 as a biomarker of renal function. The increased urinary excretion of netrin-1 during kidney disease is paralleled by a down-regulation of its plasma levels, with potential implications at a systemic level. In summary, the role of netrin-1 in cardiovascular disease is an emerging area of research requiring further in-depth study to elucidate its mechanism of action and potential as a therapeutic target, especially in view of its seemingly contradictory actions in certain physiological pathways which serve to highlight its manifold and often opposite effects in numerous physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Kerry Layne
- Cardiovascular Division, British Heart Foundation Centre for Research Excellence, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Albert Ferro
- Cardiovascular Division, British Heart Foundation Centre for Research Excellence, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Gabriella Passacquale
- Cardiovascular Division, British Heart Foundation Centre for Research Excellence, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| |
Collapse
|
83
|
Passacquale G, Phinikaridou A, Warboys C, Cooper M, Lavin B, Alfieri A, Andia ME, Botnar RM, Ferro A. Aspirin-induced histone acetylation in endothelial cells enhances synthesis of the secreted isoform of netrin-1 thus inhibiting monocyte vascular infiltration. Br J Pharmacol 2015; 172:3548-64. [PMID: 25824964 PMCID: PMC4507159 DOI: 10.1111/bph.13144] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 02/19/2015] [Accepted: 03/23/2015] [Indexed: 12/18/2022] Open
Abstract
Background and Purpose There are conflicting data regarding whether netrin-1 retards or accelerates atherosclerosis progression, as it can lead either to monocyte repulsion from or retention within plaques depending on its cellular source. We investigated the effect of aspirin, which is widely used in cardiovascular prophylaxis, on the synthesis of different isoforms of netrin-1 by endothelial cells under pro-inflammatory conditions, and defined the net effect of aspirin-dependent systemic modulation of netrin-1 on atherosclerosis progression. Experimental Approach Netrin-1 synthesis was studied in vitro using human endothelial cells stimulated with TNF-α, with or without aspirin treatment. In vivo experiments were conducted in ApoE−/− mice fed with a high-fat diet (HFD), receiving either aspirin or clopidogrel. Key Results TNF-α-induced NF-κB activation up-regulated the nuclear isoform of netrin-1, while simultaneously reducing secreted netrin-1. Down-regulation of the secreted isoform compromised the chemorepellent action of the endothelium against monocyte chemotaxis. Aspirin counteracted TNF-α-mediated effects on netrin-1 synthesis by endothelial cells through COX-dependent inhibition of NF-κB and concomitant histone hyperacetylation. Administration of aspirin to ApoE−/− mice on HFD increased blood and arterial wall levels of netrin-1 independently of its effects on platelets, accompanied by reduced plaque size and content of monocytes/macrophages, compared with untreated or clopidogrel-treated mice. In vivo blockade of netrin-1 enhanced monocyte plaque infiltration in aspirin-treated ApoE−/− mice. Conclusions and Implications Aspirin counteracts down-regulation of secreted netrin-1 induced by pro-inflammatory stimuli in endothelial cells. The aspirin-dependent increase of netrin-1 in ApoE−/− mice exerts anti-atherogenic effects by preventing arterial accumulation of monocytes.
Collapse
Affiliation(s)
- Gabriella Passacquale
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Alkystis Phinikaridou
- Division of Imaging Sciences and Biomedical Engineering, BHF Centre of Research Excellence and the Wellcome Trust/EPSRC Medical Engineering Centre, King's College London, London, UK
| | - Christina Warboys
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Margaret Cooper
- Division of Imaging Sciences and Biomedical Engineering, BHF Centre of Research Excellence and the Wellcome Trust/EPSRC Medical Engineering Centre, King's College London, London, UK
| | - Begona Lavin
- Division of Imaging Sciences and Biomedical Engineering, BHF Centre of Research Excellence and the Wellcome Trust/EPSRC Medical Engineering Centre, King's College London, London, UK
| | - Alessio Alfieri
- Department of Vascular Biology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Marcelo E Andia
- Division of Imaging Sciences and Biomedical Engineering, BHF Centre of Research Excellence and the Wellcome Trust/EPSRC Medical Engineering Centre, King's College London, London, UK
| | - Rene M Botnar
- Division of Imaging Sciences and Biomedical Engineering, BHF Centre of Research Excellence and the Wellcome Trust/EPSRC Medical Engineering Centre, King's College London, London, UK
| | - Albert Ferro
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| |
Collapse
|
84
|
Ghosh S, Vivar J, Nelson CP, Willenborg C, Segrè AV, Mäkinen VP, Nikpay M, Erdmann J, Blankenberg S, O'Donnell C, März W, Laaksonen R, Stewart AFR, Epstein SE, Shah SH, Granger CB, Hazen SL, Kathiresan S, Reilly MP, Yang X, Quertermous T, Samani NJ, Schunkert H, Assimes TL, McPherson R. Systems Genetics Analysis of Genome-Wide Association Study Reveals Novel Associations Between Key Biological Processes and Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2015; 35:1712-22. [PMID: 25977570 DOI: 10.1161/atvbaha.115.305513] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 04/28/2015] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Genome-wide association studies have identified multiple genetic variants affecting the risk of coronary artery disease (CAD). However, individually these explain only a small fraction of the heritability of CAD and for most, the causal biological mechanisms remain unclear. We sought to obtain further insights into potential causal processes of CAD by integrating large-scale GWA data with expertly curated databases of core human pathways and functional networks. APPROACHES AND RESULTS Using pathways (gene sets) from Reactome, we carried out a 2-stage gene set enrichment analysis strategy. From a meta-analyzed discovery cohort of 7 CAD genome-wide association study data sets (9889 cases/11 089 controls), nominally significant gene sets were tested for replication in a meta-analysis of 9 additional studies (15 502 cases/55 730 controls) from the Coronary ARtery DIsease Genome wide Replication and Meta-analysis (CARDIoGRAM) Consortium. A total of 32 of 639 Reactome pathways tested showed convincing association with CAD (replication P<0.05). These pathways resided in 9 of 21 core biological processes represented in Reactome, and included pathways relevant to extracellular matrix (ECM) integrity, innate immunity, axon guidance, and signaling by PDRF (platelet-derived growth factor), NOTCH, and the transforming growth factor-β/SMAD receptor complex. Many of these pathways had strengths of association comparable to those observed in lipid transport pathways. Network analysis of unique genes within the replicated pathways further revealed several interconnected functional and topologically interacting modules representing novel associations (eg, semaphoring-regulated axonal guidance pathway) besides confirming known processes (lipid metabolism). The connectivity in the observed networks was statistically significant compared with random networks (P<0.001). Network centrality analysis (degree and betweenness) further identified genes (eg, NCAM1, FYN, FURIN, etc) likely to play critical roles in the maintenance and functioning of several of the replicated pathways. CONCLUSIONS These findings provide novel insights into how genetic variation, interpreted in the context of biological processes and functional interactions among genes, may help define the genetic architecture of CAD.
Collapse
|
85
|
Affiliation(s)
- Ziad Mallat
- From the Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom; and Institut National de la Santé et de la Recherche Médicale, U970, Paris, France.
| |
Collapse
|
86
|
Tanaka J, Tanaka H, Mizuki N, Nomura E, Ito N, Nomura N, Yamane M, Hida T, Goshima Y, Hatano H, Nakagawa H. Semaphorin 3A controls allergic and inflammatory responses in experimental allergic conjunctivitis. Int J Ophthalmol 2015; 8:1-10. [PMID: 25709899 DOI: 10.3980/j.issn.2222-3959.2015.01.01] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 12/08/2014] [Indexed: 11/02/2022] Open
Abstract
AIM To assess the efficacy of topical Semaphorin-3A (SEMA3A) in the treatment of allergic conjunctivitis. METHODS Experimental allergic conjunctivitis (EAC) mice model induced by short ragweed pollen (SRW) in 4-week-old of BALB/c mice, mice were evaluated using haematoxylin and eosin (H&E) staining, immunofluorescence and light microscope photographs. Early phase took the samples in 24h after instillation and late phase took the samples between 4 to 14d after the start of treatment. The study use of topical SEMA3A (10 U, 100 U, 1000 U) eye drops and subconjunctival injection of SEMA3A with same concentration. For comparison, five types of allergy eyedrops were quantified using clinical characteristics. RESULTS Clinical score of composite ocular symptoms of the mice treated with SEMA3A were significantly decreased both in the immediate phase and the late phase compared to those treated with commercial ophthalmic formulations and non-treatment mice. SEMA3A treatment attenuates infiltration of eosinophils entering into conjunctiva in EAC mice. The score of eosinophil infiltration in the conjunctiva of SEMA3A 1000 U-treated group were significantly lower than low-concentration of SEMA3A treated groups and non-treated group. SEMA3A treatment also suppressed T-cell proliferation in vitro and decreased serum total IgE levels in EAC mice. Moreover, Treatment of SEMA3A suppressed Th2-related cytokines (IL-5, IL-13 and IL-4) and pro-inflammatory cytokines (IFN-γ, IL-17 and TNF-α) release, but increased regulatory cytokine IL-10 concentration in the conjunctiva of EAC mice. CONCLUSIONS SEMA3A as a biological agent, showed the beneficial activity in ocular allergic processes with the less damage to the intraocular tissue. It is expected that SEMA3A may be contributed in patients with a more severe spectrum of refractory ocular allergic diseases including allergic conjunctivitis in the near future.
Collapse
Affiliation(s)
- Junmi Tanaka
- Department of Ophthalmology and Visual Science, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan ; Department of Ophthalmology, Graduate School of Medicine, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hideo Tanaka
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Nobuhisa Mizuki
- Department of Ophthalmology and Visual Science, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Eiichi Nomura
- Department of Ophthalmology and Visual Science, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Norihiko Ito
- Department of Ophthalmology and Visual Science, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Naoko Nomura
- Department of Ophthalmology and Visual Science, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Masayuki Yamane
- Department of Environmental Immuno-Dermatology, Graduate School of Medicine, Yokohama City University 3-9 Fukuura, Kanazawa-ku Yokohama 236-0004 Japan
| | - Tomonobu Hida
- Department of Environmental Immuno-Dermatology, Graduate School of Medicine, Yokohama City University 3-9 Fukuura, Kanazawa-ku Yokohama 236-0004 Japan
| | - Yoshio Goshima
- Department of Environmental Immuno-Dermatology, Graduate School of Medicine, Yokohama City University 3-9 Fukuura, Kanazawa-ku Yokohama 236-0004 Japan
| | - Hiroshi Hatano
- Hatano Eye Clinic, 438-1 Fujisawa, Fujisawa, Kanagawa-ken 251-0052, Japan
| | - Hisashi Nakagawa
- Tokushima Eye Clinic, 1-2-14 Fujimi-cho, Higashimurayama, Tokyo 189-0024, Japan
| |
Collapse
|
87
|
Corà D, Astanina E, Giraudo E, Bussolino F. Semaphorins in cardiovascular medicine. Trends Mol Med 2014; 20:589-98. [PMID: 25154329 DOI: 10.1016/j.molmed.2014.07.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 07/12/2014] [Accepted: 07/23/2014] [Indexed: 01/08/2023]
Abstract
During organogenesis, patterning is primarily achieved by the combined actions of morphogens. Among these, semaphorins represent a general system for establishing the appropriate wiring architecture of biological nets. Originally discovered as evolutionarily conserved steering molecules for developing axons, subsequent studies on semaphorins expanded their functions to the cardiovascular and immune systems. Semaphorins participate in cardiac organogenesis and control physiological vasculogenesis and angiogenesis, which result from a balance between pro- and anti-angiogenic signals. These signals are altered in several diseases. In this review, we discuss the role of semaphorins in vascular biology, emphasizing the mechanisms by which these molecules control vascular patterning and lymphangiogenesis, as well as in genetically inherited and degenerative vascular diseases.
Collapse
Affiliation(s)
- Davide Corà
- Department of Oncology, University of Torino, Torino, Italy; Candiolo Cancer Institute, Torino, Candiolo, Italy
| | - Elena Astanina
- Department of Oncology, University of Torino, Torino, Italy; Candiolo Cancer Institute, Torino, Candiolo, Italy
| | - Enrico Giraudo
- Candiolo Cancer Institute-FPO, IRCCS, Torino, Candiolo, Italy; Department of Science and Drug Technology, University of Torino, Torino, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, Torino, Italy; Candiolo Cancer Institute, Torino, Candiolo, Italy.
| |
Collapse
|
88
|
Enoki Y, Sato T, Tanaka S, Iwata T, Usui M, Takeda S, Kokabu S, Matsumoto M, Okubo M, Nakashima K, Yamato M, Okano T, Fukuda T, Chida D, Imai Y, Yasuda H, Nishihara T, Akita M, Oda H, Okazaki Y, Suda T, Yoda T. Netrin-4 derived from murine vascular endothelial cells inhibits osteoclast differentiation in vitro and prevents bone loss in vivo. FEBS Lett 2014; 588:2262-9. [PMID: 24846137 DOI: 10.1016/j.febslet.2014.05.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 04/23/2014] [Accepted: 05/06/2014] [Indexed: 01/30/2023]
Abstract
Bone is a highly vascularized organ, thus angiogenesis is a vital process during bone remodeling. However, the role of vascular systems in bone remodeling is not well recognized. Here we show that netrin-4 inhibits osteoclast differentiation in vitro and in vivo. Co-cultures of bone marrow macrophages with vascular endothelial cells markedly inhibited osteoclast differentiation. Adding a neutralizing antibody, or RNA interference against netrin-4, restored in vitro osteoclast differentiation. Administration of netrin-4 prevented bone loss in an osteoporosis mouse model by decreasing the osteoclast number. We propose that vascular endothelial cells interact with bone in suppressing bone through netrin-4.
Collapse
Affiliation(s)
- Yuichiro Enoki
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan
| | - Tsuyoshi Sato
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan.
| | - Shinya Tanaka
- Department of Orthopedic Surgery, Saitama Medical University, Saitama, Japan
| | - Takanori Iwata
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Michihiko Usui
- Department of Periodontology, Kyushu Dental University, Fukuoka, Japan
| | - Shu Takeda
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shoichiro Kokabu
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan
| | - Masahito Matsumoto
- Division of Functional Genomics and Systems Medicine, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Masahiko Okubo
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan; Division of Functional Genomics and Systems Medicine, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Keisuke Nakashima
- Department of Periodontology, Kyushu Dental University, Fukuoka, Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Toru Fukuda
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Dai Chida
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan
| | - Yuuki Imai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Ehime, Japan
| | - Hisataka Yasuda
- Planning & Development, Bioindustry Division, Oriental Yeast Co., Ltd., Tokyo, Japan
| | - Tatsuji Nishihara
- Division of Infections and Molecular Biology, Kyushu Dental University, Fukuoka, Japan
| | - Masumi Akita
- Division of Morphological Science, Saitama Medical University, Saitama, Japan
| | - Hiromi Oda
- Department of Orthopedic Surgery, Saitama Medical University, Saitama, Japan
| | - Yasushi Okazaki
- Division of Functional Genomics and Systems Medicine, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Tatsuo Suda
- Division of Functional Genomics and Systems Medicine, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Tetsuya Yoda
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan
| |
Collapse
|
89
|
Taghavie-Moghadam PL, Butcher MJ, Galkina EV. The dynamic lives of macrophage and dendritic cell subsets in atherosclerosis. Ann N Y Acad Sci 2014; 1319:19-37. [PMID: 24628328 DOI: 10.1111/nyas.12392] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Atherosclerosis, the major pathological process through which arterial plaques are formed, is a dynamic chronic inflammatory disease of large- and medium-sized arteries in which the vasculature, lipid metabolism, and the immune system all play integral roles. Both the innate and adaptive immune systems are involved in the development and progression of atherosclerosis but myeloid cells represent the major component of the burgeoning atherosclerotic plaque. Various myeloid cells, including monocytes, macrophages (MΦs), and dendritic cells (DCs) can be found within the healthy and atherosclerotic arterial wall, where they can contribute to or regulate inflammation. However, the precise behaviors and functions of these cells in situ are still active areas of investigation that continue to yield exciting and surprising new data. Here, we review recent progress in understanding of the complex biology of MΦs and DCs, focusing particularly on the dynamic regulation of these subsets in the arterial wall and novel, emerging functions of these cells during atherogenesis.
Collapse
Affiliation(s)
- Parésa L Taghavie-Moghadam
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia
| | | | | |
Collapse
|
90
|
Oksala N, Pärssinen J, Seppälä I, Raitoharju E, Ivana K, Hernesniemi J, Lyytikäinen LP, Levula M, Mäkelä KM, Sioris T, Kähönen M, Laaksonen R, Hytönen V, Lehtimäki T. Association of Neuroimmune Guidance Cue Netrin-1 and Its Chemorepulsive Receptor UNC5B With Atherosclerotic Plaque Expression Signatures and Stability in Human(s). ACTA ACUST UNITED AC 2013; 6:579-87. [DOI: 10.1161/circgenetics.113.000141] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background—
Macrophage (MΦ) infiltration and smooth muscle cell (SMC) proliferation are hallmarks of atherosclerosis and unstable plaques. Neuroimmune guidance cue 1 (netrin-1 [NTN1]) plays a critical role controlling MΦ trafficking and SMC activation. Characterization of expression of NTN1 and its receptors and their association with plaque stability in human(s) is lacking.
Methods and Results—
The expression of NTN1 and its receptors did not differ in either whole blood or circulating monocytes from patients with coronary artery disease (n=55) compared with healthy controls (n=45). However, NTN1 was downregulated (−2.9-fold;
P
<0.0001) and UNC5B upregulated (2.2-fold;
P
<0.0001) in atherosclerotic plaques (n=68), whereas there were no differences in other NTN1 receptors compared with histologically normal controls (n=28). Increased UNC5B expression is associated with histologically more stable plaques (
P
=0.011). NTN1 expression correlated positively with SMC markers and signatures and negatively with inflammatory markers and M1 and especially M2 signatures in the atherosclerotic plaques. UNC5B clustering correlated positively with inflammatory and MΦ markers. NTN1 protein colocalized with CD68-positive cells of monocytic origin and muscle-actin-specific-antibody (HHF3)-positive cells indicative of SMCs in the plaques and only with SMCs in the control samples. NTN1 protein was highly expressed in the intimal layer of the control vessels.
Conclusions—
Present findings provide support for the hypothesis that dysregulation of expression of NTN1 in SMCs and its chemorepulsive receptor UNC5B in macrophages are involved in the development of atherosclerosis and unstable plaques.
Collapse
Affiliation(s)
- Niku Oksala
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Jenita Pärssinen
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Ilkka Seppälä
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Emma Raitoharju
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Kholova Ivana
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Jussi Hernesniemi
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Mari Levula
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Kari-Matti Mäkelä
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Thanos Sioris
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Mika Kähönen
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Reijo Laaksonen
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Vesa Hytönen
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Terho Lehtimäki
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| |
Collapse
|
91
|
Abstract
Atherosclerosis is a chronic inflammatory disease that arises from an imbalance in lipid metabolism and a maladaptive immune response driven by the accumulation of cholesterol-laden macrophages in the artery wall. Through the analysis of the progression and regression of atherosclerosis in animal models, there is a growing understanding that the balance of macrophages in the plaque is dynamic and that both macrophage numbers and the inflammatory phenotype influence plaque fate. In this Review, we summarize recently identified pro- and anti-inflammatory pathways that link lipid and inflammation biology with the retention of macrophages in plaques, as well as factors that have the potential to promote their egress from these sites.
Collapse
|
92
|
Randolph GJ, Gautier EL. Emerging roles of neural guidance molecules in atherosclerosis: sorting out the complexity. Arterioscler Thromb Vasc Biol 2013; 33:882-3. [PMID: 23576713 DOI: 10.1161/atvbaha.113.301346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
93
|
Ramkhelawon B, Yang Y, van Gils JM, Hewing B, Rayner KJ, Parathath S, Guo L, Oldebeken S, Feig JL, Fisher EA, Moore KJ. Hypoxia induces netrin-1 and Unc5b in atherosclerotic plaques: mechanism for macrophage retention and survival. Arterioscler Thromb Vasc Biol 2013; 33:1180-8. [PMID: 23599441 DOI: 10.1161/atvbaha.112.301008] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Hypoxia is intimately linked to atherosclerosis and has become recognized as a primary impetus of inflammation. We recently demonstrated that the neuroimmune guidance cue netrin-1 (Ntn1) inhibits macrophage emigration from atherosclerotic plaques, thereby fostering chronic inflammation. However, the mechanisms governing netrin-1 expression in atherosclerosis are not well understood. In this study, we investigate the role of hypoxia in regulating expression of netrin-1 and its receptor uncoordinated-5-B receptor (Unc5b) in plaque macrophages and its functional consequences on these immune cells. APPROACH AND RESULTS We show by immunostaining that netrin-1 and Unc5b are expressed in macrophages in hypoxia-rich regions of human and mouse plaques. In vitro, Ntn1 and Unc5b mRNA are upregulated in macrophages treated with oxidized low-density lipoprotein or inducers of oxidative stress (CoCl2, dimethyloxalylglycine, 1% O2). These responses are abrogated by inhibiting hypoxia-inducible transcription factor (HIF)-1α, indicating a causal role for this transcription factor in regulating Ntn1 and Unc5b expression in macrophages. Indeed, using promoter-luciferase reporter genes, we show that Ntn1- and Unc5b-promoter activities are induced by oxidized low-density lipoprotein and require HIF-1α. Correspondingly, J774 macrophages overexpressing active HIF-1α show increased netrin-1 and Unc5b expression and reduced migratory capacity compared with control cells, which was restored by blocking the effects of netrin-1. Finally, we show that netrin-1 protects macrophages from apoptosis under hypoxic conditions in a HIF-1α-dependent manner. CONCLUSIONS These findings provide a molecular mechanism by which netrin-1 and its receptor Unc5b are expressed in atherosclerotic plaques and implicate hypoxia and HIF-1α-induced netrin-1/Unc5b in sustaining inflammation by inhibiting the emigration and promoting the survival of lesional macrophages.
Collapse
Affiliation(s)
- Bhama Ramkhelawon
- Department of Medicine, Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|