1
|
Kant S, Caliz AD, Yoo HJ, Kaur G, Learnard H, Khalil HA, Davis RJ, Keaney JFJ. Mixed lineage kinase (MLK) controls tumor development and angiogenesis. Angiogenesis 2025; 28:29. [PMID: 40314847 DOI: 10.1007/s10456-025-09978-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025]
Abstract
Cancer is among the leading causes of death in the USA and worldwide. Solid tumors require the formation of new blood vessels (angiogenesis) for their growth. The endothelium plays a crucial role in angiogenesis and tumor progression. Hypoxic stress generated by tumors can activate stress kinases such as mixed lineage kinases (MLKs). Publicly available datasets on lung adenocarcinoma, along with our experimental findings, indicate that MLK2 and MLK3 are expressed in human lung tumors. In this study, using three distinct mouse models of tumor development, we demonstrated that MLK2 (MAP3K10) and MLK3 (MAP3K11) are essential for tumor growth and angiogenesis. Furthermore, MLK2 and MLK3 are highly expressed in the endothelium and are necessary for endothelial proliferation, migration, and angiogenesis. In the endothelium, MLKs regulate the expression of angiogenic growth factors and metalloproteinases, including Pgf, Vegfa, Angptl4, Adam8, and Mmp9. Additionally, the MLK family of kinases acts through the long noncoding RNA (lncRNA) H19 to control the expression of these pro-angiogenic factors in the endothelium. Collectively, these findings suggest that the MLK-H19 axis coordinates endothelial function, angiogenesis, and tumor growth.
Collapse
Affiliation(s)
- Shashi Kant
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Amada D Caliz
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hyung-Jin Yoo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Gaganpreet Kaur
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Heather Learnard
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Hassan A Khalil
- Division of Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Roger J Davis
- Program of Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - John F Jr Keaney
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
2
|
Orfanidou M, Polyzos SA. Retinopathy in Metabolic Dysfunction-Associated Steatotic Liver Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 61:38. [PMID: 39859020 PMCID: PMC11766779 DOI: 10.3390/medicina61010038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a multisystemic disease, i.e., influencing various organ systems beyond the liver and, thus, contributing to comorbidities. Characterized by excessive fat accumulation in the hepatocytes, MASLD is frequently linked to metabolic syndrome components, such as obesity, insulin resistance, dyslipidemia, and hypertension. Therefore, exploring the intricate connection between MASLD and other organ systems, including the eyes, seems to be essential. In this context, retinopathy has been investigated for its potential association with MASLD, since both conditions share common pathogenetic pathways. Chronic low-grade inflammation, oxidative stress, insulin resistance, and endothelial dysfunction are only some of those mechanisms contributing to disease progression and, possibly, determining the bidirectional interplay between the liver and retinal pathology. This narrative review aims to summarize data concerning the multisystemicity of MASLD, primarily focusing on its potential association with the eyes and, particularly, retinopathy. Identifying this possible association may emphasize the need for early screening and integrated management approaches that address the liver and eyes as interconnected components within the framework of a systemic disease. Further research is necessary to delineate the precise mechanisms and develop targeted interventions to mitigate the bidirectional impact between the liver and eyes, aiming to reduce the overall burden of disease and improve patient outcomes.
Collapse
Affiliation(s)
- Myrsini Orfanidou
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- First Department of Ophthalmology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Stergios A. Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
3
|
Lu X, Fan Z, Yang S, Zheng L, Yu Z, Yang Y, Zheng M, Zeng J, Zhang G. Role of angiopoietin-like 4 in neovascularization associated with retinopathy of prematurity. Exp Eye Res 2024; 249:110145. [PMID: 39491782 DOI: 10.1016/j.exer.2024.110145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/08/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
To elucidate the mechanisms of angiopoietin-like 4 (ANGTPL4) in neovascularization (NV) in retinopathy of prematurity (ROP). We compared ANGPTL4 expression levels of aqueous humour and vitreous fluid samples in infants with acute-phase ROP and control group. ANGPTL4-/- mice and WT mice were used to constructed oxygen-induced retinopathy (OIR) mouse models, with retinal tissues collected on postnatal days 12 (P12), 15 (P15) and 17 (P17). Analysis of retinal vessels and transcriptomics were performed to explore the role of ANGTPL4 in NV. The results showed ANGPTL4 level was significantly higher in the aqueous humour and vitreous fluid of children with ROP than that of control group. At P15 and P17, the vascular indices in the ANGPTL4-/--CON group were lower than those in the WT-CON group. The central non-perfused area of the retina and number of neovascular nuclei were also smaller in the ANGPTL4-/--OIR group than in the WT-OIR group. Immunofluorescence results showed the overexpression of ANGPTL4 protein in the WT-OIR group than in the WT-CON group, especially at P17. Furthermore, extracellular matrix (ECM) organisation was one of the key involved pathways based on gene ontology (GO) enrichment analyses. ANGPTL4 was one of the core genes involved in ECM organization, and neuralized E3 ubiquitin protein ligase 1B (NEURL1B), cd36 Molecule (CD36), matrix metallopeptidase 3 (MMP3) and collagen type III alpha 1 chain (COL3A1) were the first nodes interacting with ANGPTL4.In conclusion, ANGPTL4 is involved in the pathological NV by regulating NEURL1B, CD36, MMP3, and COL3A1. Thus, ANGPTL4 is a potential therapeutic target for ROP.
Collapse
Affiliation(s)
- Xiaofeng Lu
- Department of Pediatric Retinal Surgery, Shenzhen Eye Hospital, 18 Zetian Road, Shenzhen 518040, Guangdong Province, China
| | - Zixin Fan
- Department of Pediatric Retinal Surgery, Shenzhen Eye Hospital, 18 Zetian Road, Shenzhen 518040, Guangdong Province, China
| | - Shuo Yang
- Department of Pediatric Retinal Surgery, Shenzhen Eye Hospital, 18 Zetian Road, Shenzhen 518040, Guangdong Province, China
| | - Lei Zheng
- Department of Pediatric Retinal Surgery, Shenzhen Eye Hospital, 18 Zetian Road, Shenzhen 518040, Guangdong Province, China
| | - Zhen Yu
- Department of Pediatric Retinal Surgery, Shenzhen Eye Hospital, 18 Zetian Road, Shenzhen 518040, Guangdong Province, China
| | - Yuhang Yang
- Department of Pediatric Retinal Surgery, Shenzhen Eye Hospital, 18 Zetian Road, Shenzhen 518040, Guangdong Province, China
| | - Mianying Zheng
- Department of Pediatric Retinal Surgery, Shenzhen Eye Hospital, 18 Zetian Road, Shenzhen 518040, Guangdong Province, China
| | - Jian Zeng
- Department of Pediatric Retinal Surgery, Shenzhen Eye Hospital, 18 Zetian Road, Shenzhen 518040, Guangdong Province, China
| | - Guoming Zhang
- Department of Pediatric Retinal Surgery, Shenzhen Eye Hospital, 18 Zetian Road, Shenzhen 518040, Guangdong Province, China.
| |
Collapse
|
4
|
Wang Y, Zhang Y, Qu Y, Li S, Xi W, Liu B, Ye L. eIF4A3-mediated circEHMT1 regulation in retinal microvascular endothelial dysfunction in diabetic retinopathy. Microvasc Res 2024; 151:104612. [PMID: 37839527 DOI: 10.1016/j.mvr.2023.104612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND AND OBJECTIVE Literature has reported that circular RNAs (circRNAs) are crucially associated with diabetic retinopathy (DR). Furthermore, circEHMT1 has been identified to maintain endothelial cell barrier function. This study aimed to investigate the mechanisms that regulate aberrant circEHMT1 expression and its role in the pathogenesis of DR. METHODS In this study, retinal microvascular endothelial cells were exposed to a high glucose (HG) environment, and subsequently, tube formation and intercellular junction proteins were evaluated. Furthermore, the biological functions of circEHMT1 and its potential regulatory factor, eIF4A3, in microvascular endothelial cells under HG conditions were also assessed. In addition, the regulatory role of eIF4A3 on circEHMT1 expression was confirmed. Moreover, to elucidate the in vivo functions of eIF4A3 and circEHMT1, streptozotocin (STZ) was used to establish a DR model in rats. RESULTS It was revealed that HG condition decreased circEHMT1 and eIF4A3 expressions and reduced ZO-1, Claudin-5, and Occludin levels in retinal microvascular endothelial cells. Furthermore, it was observed that eIF4A3 could regulate the expression of circEHMT1. Overexpression of eIF4A3 or circEHMT1 under HG conditions improved endothelial cell injury and decreased tube-formation ability. Additionally, in the DR rat model, eIF4A3 overexpression restored circEHMT1 levels and ameliorated retinal vasculature changes. CONCLUSION Altogether, eIF4A3 regulates circEHMT1 expression, thereby affecting microvascular endothelial cell injury and tube formation. Further understanding the regulatory effect of eIF4A3 on circEHMT1 may provide novel therapeutic targets for DR.
Collapse
Affiliation(s)
- Yuan Wang
- Shenzhen Eye Hospital, Jinan University, Shenzhen 518040, China; Shenzhen Eye Institute, Shenzhen 518040, China
| | - Yongxin Zhang
- Shenzhen Eye Hospital, Jinan University, Shenzhen 518040, China; Shenzhen Eye Institute, Shenzhen 518040, China
| | - Yunhao Qu
- Shenzhen Eye Hospital, Jinan University, Shenzhen 518040, China
| | - Shixu Li
- Shenzhen Eye Hospital, Jinan University, Shenzhen 518040, China
| | - Wenqun Xi
- Shenzhen Eye Hospital, Jinan University, Shenzhen 518040, China; Shenzhen Eye Institute, Shenzhen 518040, China
| | - Beian Liu
- Shenzhen Eye Hospital, Jinan University, Shenzhen 518040, China
| | - Lin Ye
- Shenzhen Eye Hospital, Jinan University, Shenzhen 518040, China; Shenzhen Eye Institute, Shenzhen 518040, China.
| |
Collapse
|
5
|
Chaube B, Citrin KM, Sahraei M, Singh AK, de Urturi DS, Ding W, Pierce RW, Raaisa R, Cardone R, Kibbey R, Fernández-Hernando C, Suárez Y. Suppression of angiopoietin-like 4 reprograms endothelial cell metabolism and inhibits angiogenesis. Nat Commun 2023; 14:8251. [PMID: 38086791 PMCID: PMC10716292 DOI: 10.1038/s41467-023-43900-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Angiopoietin-like 4 (ANGPTL4) is known to regulate various cellular and systemic functions. However, its cell-specific role in endothelial cells (ECs) function and metabolic homeostasis remains to be elucidated. Here, using endothelial-specific Angptl4 knock-out mice (Angptl4iΔEC), and transcriptomics and metabolic flux analysis, we demonstrate that ANGPTL4 is required for maintaining EC metabolic function vital for vascular permeability and angiogenesis. Knockdown of ANGPTL4 in ECs promotes lipase-mediated lipoprotein lipolysis, which results in increased fatty acid (FA) uptake and oxidation. This is also paralleled by a decrease in proper glucose utilization for angiogenic activation of ECs. Mice with endothelial-specific deletion of Angptl4 showed decreased pathological neovascularization with stable vessel structures characterized by increased pericyte coverage and reduced permeability. Together, our study denotes the role of endothelial-ANGPTL4 in regulating cellular metabolism and angiogenic functions of EC.
Collapse
Affiliation(s)
- Balkrishna Chaube
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA
| | - Kathryn M Citrin
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
| | - Mahnaz Sahraei
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Abhishek K Singh
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Diego Saenz de Urturi
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA
| | - Wen Ding
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Richard W Pierce
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Raaisa Raaisa
- Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Rebecca Cardone
- Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Richard Kibbey
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
- Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Carlos Fernández-Hernando
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Yajaira Suárez
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA.
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
6
|
Gomes C, Varo R, Duran-Frigola M, Sitoe A, Bila R, Machevo S, Mayor A, Bassat Q, Rodriguez A. Endothelial transcriptomic analysis identifies biomarkers of severe and cerebral malaria. JCI Insight 2023; 8:e172845. [PMID: 37788095 PMCID: PMC10721316 DOI: 10.1172/jci.insight.172845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/28/2023] [Indexed: 10/05/2023] Open
Abstract
Malaria can quickly progress from an uncomplicated infection into a life-threatening severe disease. However, the unspecificity of early symptoms often makes it difficult to identify patients at high risk of developing severe disease. Additionally, one of the most feared malaria complications - cerebral malaria - is challenging to diagnose, often resulting in treatment delays that can lead to adverse outcomes. To identify candidate biomarkers for the prognosis and/or diagnosis of severe and cerebral malaria, we have analyzed the transcriptomic response of human brain microvascular endothelial cells to erythrocytes infected with Plasmodium falciparum. Candidates were validated in plasma samples from a cohort of pediatric patients with malaria from Mozambique, resulting in the identification of several markers with capacity to distinguish uncomplicated from severe malaria, the most potent being the metallopeptidase ADAMTS18. Two other biomarkers, Angiopoietin-like-4 and Inhibin-βE were able to differentiate children with cerebral malaria within the severe malaria group, showing increased sensitivity after combination in a biomarker signature. The validation of the predicted candidate biomarkers in plasma of children with severe and cerebral malaria underscores the power of this transcriptomic approach and indicates that a specific endothelial response to P. falciparum-infected erythrocytes is linked to the pathophysiology of severe malaria.
Collapse
Affiliation(s)
- Cláudia Gomes
- New York University Grossman School of Medicine, New York, USA
| | - Rosauro Varo
- ISGlobal, Hospital Clínic — University of Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | | | - Antonio Sitoe
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Rubão Bila
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Sonia Machevo
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Alfredo Mayor
- ISGlobal, Hospital Clínic — University of Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Carlos III Health Institute, Madrid, Spain
- Department of Physiologic Sciences, Faculty of Medicine, Universidade Eduardo Mondlane, Maputo, Mozambique
| | - Quique Bassat
- ISGlobal, Hospital Clínic — University of Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Carlos III Health Institute, Madrid, Spain
- ICREA, Pg. Lluís Companys 23, Barcelona, Spain
- Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Barcelona, Spain
| | - Ana Rodriguez
- New York University Grossman School of Medicine, New York, USA
| |
Collapse
|
7
|
Jin Z, Guo Q, Wang Z, Wu X, Hu W, Li J, Li H, Zhu S, Zhang H, Chen Z, Xu H, Shi L, Yang L, Wang Y. Andrographolide suppresses hypoxia-induced embryonic hyaloid vascular system development through HIF-1a/VEGFR2 signaling pathway. Front Cardiovasc Med 2023; 10:1090938. [PMID: 36844722 PMCID: PMC9944699 DOI: 10.3389/fcvm.2023.1090938] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/04/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction Ocular abnormalities and the development of retinal vasculature may cause postnatal retinopathy. In the past decade, tremendous progress has been made in identifying the mechanisms that regulate retina vasculature. However, the means of regulating embryonic hyaloid vasculature development is largely unknown. This study aims to determine whether and how andrographolide regulates embryonic hyaloid vasculature development. Methods Murine embryonic retinas were used in this study. Whole mount isolectin B4 (IB4) staining, hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC), and immunofluorescence staining (IF) were performed to determine whether andrographolide is critical for embryonic hyaloid vasculature development. BrdU incorporation assay, Boyden chamber migration assay, spheroid sprouting assay, and Matrigel-based tube formation assay were performed to evaluate whether andrographolide regulates the proliferation and migration of vascular endothelial cells. Molecular docking simulation and Co-immunoprecipitation assay were used to observe protein interaction. Results Hypoxia conditions exist in murine embryonic retinas. Hypoxia induces HIF-1a expression; high-expressed HIF-1a interacts with VEGFR2, resulting in the activation of the VEGF signaling pathway. Andrographolide suppresses hypoxia-induced HIF-1a expression and, at least in part, interrupts the interaction between HIF-1a and VEGFR2, causing inhibiting endothelial proliferation and migration, eventually inhibiting embryonic hyaloid vasculature development. Conclusion Our data demonstrated that andrographolide plays a critical role in regulating embryonic hyaloid vasculature development.
Collapse
Affiliation(s)
- Zhong Jin
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiru Guo
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zheng Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Wu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wangming Hu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiali Li
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongfei Li
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Song Zhu
- Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haidi Zhang
- Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zixian Chen
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Xu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liangqin Shi
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lan Yang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China,*Correspondence: Yong Wang, ✉ ; ✉
| |
Collapse
|
8
|
Zhang L, Han Y, Chen Q, Dissanayaka WL. Sema4D-plexin-B1 signaling in recruiting dental stem cells for vascular stabilization on a microfluidic platform. LAB ON A CHIP 2022; 22:4632-4644. [PMID: 36331411 DOI: 10.1039/d2lc00632d] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The recruitment of mural cells is critical for stabilization of nascent vessels. Stem cells from human exfoliated deciduous teeth (SHED) are considered to have mural cell-like properties. However, the signaling mechanisms that regulate the cross-talk between endothelial cells and SHED in recruiting them as mural cells is much less well understood. Herein, using a 3D biomimetic microfluidic device, for the first time, we unraveled the role of semaphorin 4D (Sema4D)-plexin-B1 signaling in the recruitment of SHED as mural cells during angiogenic sprouting and vasculature formation by endothelial cells (ECs) in a 3D fibrin matrix. The specific compartmentalized design of the microfluidic chip facilitated recreation of the multi-step dynamic process of angiogenesis in a time and space dependent manner. Enabled by the chip design, different morphogenic steps of angiogenesis including endothelial proliferation, migration & invasion, vascular sprout formation and recruitment of mural cells as well as functional aspects including perfusion and permeability were examined under various pharmacological and genetic manipulations. The results showed that Sema4D facilitates the interaction between endothelial cells and SHED and promotes the recruitment of SHED as mural cells in vascular stabilization. Our results further demonstrated that Sema4D exerts these effects by acting on endothelial-plexin-B1 by inducing expression of platelet-derived growth factor (PDGF)-BB, which is a major mural cell recruitment factor.
Collapse
Affiliation(s)
- Lili Zhang
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR.
| | - Yuanyuan Han
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR.
| | - Qixin Chen
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR.
| | - Waruna Lakmal Dissanayaka
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
9
|
Son Y, Paton CM. A Review of free fatty acid-induced cell signaling, angiopoietin-like protein 4, and skeletal muscle differentiation. Front Physiol 2022; 13:987977. [PMID: 36148297 PMCID: PMC9485487 DOI: 10.3389/fphys.2022.987977] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Postnatal skeletal muscle differentiation from quiescent satellite cells is a highly regulated process, although our understanding of the contribution of nutritional factors in myogenesis is limited. Free fatty acids (FFAs) are known to cause detrimental effects to differentiated skeletal muscle cells by increasing oxidative stress which leads to muscle wasting and insulin resistance in skeletal muscle. In addition, FFAs are thought to act as inhibitors of skeletal muscle differentiation. However, the precise molecular mechanisms underlying the effects of FFAs on skeletal muscle differentiation remains to be elucidated. There is a clear relationship between dietary FFAs and their ability to suppress myogenesis and we propose the hypothesis that the FFA-mediated increase in angiopoietin-like protein 4 (ANGPTL4) may play a role in the inhibition of differentiation. This review discusses the role of FFAs in skeletal muscle differentiation to-date and proposes potential mechanisms of FFA-induced ANGPTL4 mediated inhibition of skeletal muscle differentiation.
Collapse
Affiliation(s)
- Yura Son
- Department Nutritional Sciences, Athens, GA, United States
| | - Chad M. Paton
- Department Nutritional Sciences, Athens, GA, United States
- Department of Food Science and Technology, University of Georgia, Athens, GA, United States
- *Correspondence: Chad M. Paton,
| |
Collapse
|
10
|
Sun R, Xu Z, Zhu C, Chen T, Muñoz LE, Dai L, Zhao Y. Alpha-1 antitrypsin in autoimmune diseases: Roles and therapeutic prospects. Int Immunopharmacol 2022; 110:109001. [PMID: 35803133 DOI: 10.1016/j.intimp.2022.109001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 02/05/2023]
Abstract
Alpha-1 antitrypsin (A1AT) is a protease inhibitor in the serum. Its primary function is to inhibit the activity of a series of proteases, including proteinase 3, neutrophil elastase, metalloproteases, and cysteine-aspartate proteases. In addition, A1AT also has anti-inflammatory, anti-apoptotic, anti-oxidative stress, anti-viral, and anti-bacterial activities and plays essential roles in the regulation of tissue repair and lymphocyte differentiation and activation. The overactivation of the immune system characterizes the pathogenesis of autoimmune diseases. A1AT treatment shows beneficial effects on patients and animal models with autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematosus. This review summarizes the functions and therapeutic prospects of A1AT in autoimmune diseases.
Collapse
Affiliation(s)
- Rui Sun
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiqiang Xu
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics and Department of General Practice, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chenxi Zhu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tao Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Luis E Muñoz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lunzhi Dai
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics and Department of General Practice, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
11
|
Liabotis A, Ardidie-Robouant C, Mailly P, Besbes S, Gutierrez C, Atlas Y, Muller L, Germain S, Monnot C. Angiopoietin-like 4-Induced 3D Capillary Morphogenesis Correlates to Stabilization of Endothelial Adherens Junctions and Restriction of VEGF-Induced Sprouting. Biomedicines 2022; 10:biomedicines10020206. [PMID: 35203415 PMCID: PMC8869696 DOI: 10.3390/biomedicines10020206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 11/22/2022] Open
Abstract
Angiopoietin-like 4 (ANGPTL4) is a target of hypoxia that accumulates in the endothelial extracellular matrix. While ANGPTL4 is known to regulate angiogenesis and vascular permeability, its context-dependent role related to vascular endothelial growth factor (VEGF) has been suggested in capillary morphogenesis. We here thus develop in vitro 3D models coupled to imaging and morphometric analysis of capillaries to decipher ANGPTL4 functions either alone or in the presence of VEGF. ANGPTL4 induces the formation of barely branched and thin endothelial capillaries that display linear adherens junctions. However, ANGPTL4 counteracts VEGF-induced formation of abundant ramified capillaries presenting cell–cell junctions characterized by VE-cadherin containing reticular plaques and serrated structures. We further deciphered the early angiogenesis steps regulated by ANGPTL4. During the initial activation of endothelial cells, ANGPTL4 alone induces cell shape changes but limits the VEGF-induced cell elongation and unjamming. In the growing sprout, ANGPTL4 maintains cohesive VE-cadherin pattern and sustains moderate 3D cell migration but restricts VEGF-induced endothelium remodeling and cell migration. This effect is mediated by differential short- and long-term regulation of P-Y1175-VEGFR2 and ERK1-2 signaling by ANGPTL4. Our in vitro 3D models thus provide the first evidence that ANGPTL4 induces a specific capillary morphogenesis but also overcomes VEGF effect.
Collapse
Affiliation(s)
- Athanasia Liabotis
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, F-75005 Paris, France; (A.L.); (C.A.-R.); (P.M.); (S.B.); (C.G.); (Y.A.); (L.M.)
- Collège Doctoral, Sorbonne Université, F-75006 Paris, France
| | - Corinne Ardidie-Robouant
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, F-75005 Paris, France; (A.L.); (C.A.-R.); (P.M.); (S.B.); (C.G.); (Y.A.); (L.M.)
| | - Philippe Mailly
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, F-75005 Paris, France; (A.L.); (C.A.-R.); (P.M.); (S.B.); (C.G.); (Y.A.); (L.M.)
| | - Samaher Besbes
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, F-75005 Paris, France; (A.L.); (C.A.-R.); (P.M.); (S.B.); (C.G.); (Y.A.); (L.M.)
| | - Charly Gutierrez
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, F-75005 Paris, France; (A.L.); (C.A.-R.); (P.M.); (S.B.); (C.G.); (Y.A.); (L.M.)
| | - Yoann Atlas
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, F-75005 Paris, France; (A.L.); (C.A.-R.); (P.M.); (S.B.); (C.G.); (Y.A.); (L.M.)
- Collège Doctoral, Sorbonne Université, F-75006 Paris, France
| | - Laurent Muller
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, F-75005 Paris, France; (A.L.); (C.A.-R.); (P.M.); (S.B.); (C.G.); (Y.A.); (L.M.)
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, F-75005 Paris, France; (A.L.); (C.A.-R.); (P.M.); (S.B.); (C.G.); (Y.A.); (L.M.)
- Correspondence: (S.G.); (C.M.)
| | - Catherine Monnot
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, F-75005 Paris, France; (A.L.); (C.A.-R.); (P.M.); (S.B.); (C.G.); (Y.A.); (L.M.)
- Correspondence: (S.G.); (C.M.)
| |
Collapse
|
12
|
Yan J, Li WJ, Qin YZ, Qiu XY, Qin L, Li JM. Aqueous angiopoietin-like levels correlate with optical coherence tomography angiography metrics in diabetic macular edema. Int J Ophthalmol 2021; 14:1888-1894. [PMID: 34926204 DOI: 10.18240/ijo.2021.12.12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 09/03/2021] [Indexed: 11/23/2022] Open
Abstract
AIM To quantitatively detect aqueous levels of angiopoietin-like (ANGPTL)3, ANGPTL4, and ANGPTL6 and investigate their correlation with optical coherence tomography angiography (OCTA) findings in patients with diabetic macular edema (DME). METHODS This cross-sectional study included 23 patients (27 eyes) with type 2 diabetes and 16 control subjects (20 eyes). All patients underwent OCTA imaging and ultra-wide field fundus photography. Diabetic patients were categorized into two groups according to the presence or absence of diabetic retinopathy (DME group, 14 patients, 16 eyes); and non-diabetic retinopathy (NDR) group, 9 patients, 11 eyes, respectively. Aqueous levels of ANGPTL3, ANGPTL4, and ANGPTL6 were assessed using suspension array technology, and foveal-centered 3×3 mm2 OCTA scans were automatically graded to determine the central, inner, and full vessel density (CVD, IVD, FVD); central, inner, and full perfusion density (CPD, IPD, FPD), foveal avascular zone (FAZ) area, FAZ perimeter, and FAZ circularity index (FAZ-CI) on superficial capillary plexuses. Additionally, central subfield thickness (CST), cube volume (CV), and cube average thickness (CAT) were measured in a model of macular cube 512×128. RESULTS Aqueous ANGPTL3 levels were not significantly different among the three groups (P>0.05). ANGPTL4 levels were significantly higher in the DME group than the control and NDR groups (P<0.0001 and P<0.001), while ANGPTL6 levels were significantly higher in the DME group than the control group (P<0.05). In the whole cohort, the aqueous ANGPTL3 levels correlated negatively with the IVD, FVD, IPD, and FPD, and positively with the CV and CAT. The aqueous ANGPTL4 levels correlated negatively with the CVD, IVD, FVD, CPD, IPD, and FPD, and positively with the FAZ perimeter, CST, CV, and CAT. The aqueous ANGPTL6 levels correlated negatively with the IVD, FVD, IPD, FPD, FAZ-CI and positively with CST, CV, CAT. CONCLUSION ANGPTL4 and ANGPTL6 may be associated with vascular leakage in DME and may represent good targets for DME therapy. In addition, OCTA metrics may be useful for evaluating macular ischemia in DME.
Collapse
Affiliation(s)
- Jie Yan
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China.,Department of Ophthalmology, Yulin Hospital of Traditional Chinese Medicine, Yulin 719000, Shaanxi Province, China
| | - Wu-Jun Li
- Department of Ophthalmology, Yulin Hospital of Traditional Chinese Medicine, Yulin 719000, Shaanxi Province, China
| | - Ya-Zhou Qin
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Xuan-Yu Qiu
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Li Qin
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Jing-Ming Li
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW Since the first discovery of Angiopoetin-like 4 (ANGPTL4) in 2000, the involvement of ANGPTL4 in different aspects of lipid metabolism and vascular biology has emerged as an important research field. In this review, we summarize the fundamental roles of ANGPTL4 in regulating metabolic and nonmetabolic functions and their implication in lipid metabolism and with several aspects of vascular function and dysfunction. RECENT FINDINGS ANGPTL4 is a secreted glycoprotein with a physiological role in lipid metabolism and a predominant expression in adipose tissue and liver. ANGPTL4 inhibits the activity of lipoprotein lipase and thereby promotes an increase in circulating triglyceride levels. Therefore, ANGPTL4 has been highly scrutinized as a potential therapeutic target. Further involvement of ANGPTL4 has been shown to occur in tumorigenesis, angiogenesis, vascular permeability and stem cell regulation, which opens new opportunities of using ANGPTL4 as potential therapeutic targets for other pathophysiological conditions. SUMMARY Further determination of ANGPTL4 regulatory circuits and defining specific molecular events that mediate its biological effects remain key to future ANGPTL4-based therapeutic applications in different disease settings. Many new and unanticipated roles of ANGPTL4 in the control of cell-specific functions will assist clinicians and researchers in developing potential therapeutic applications.
Collapse
|
14
|
Qi K, Yang Y, Geng Y, Cui H, Li X, Jin C, Chen G, Tian X, Meng X. Tongxinluo attenuates oxygen-glucose-serum deprivation/restoration-induced endothelial barrier breakdown via peroxisome proliferator activated receptor-α/angiopoietin-like 4 pathway in high glucose-incubated human cardiac microvascular endothelial cells. Medicine (Baltimore) 2020; 99:e21821. [PMID: 32846824 PMCID: PMC7447398 DOI: 10.1097/md.0000000000021821] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Traditional Chinese medicine Tongxinluo (TXL) has been widely used to treat coronary artery disease in China, since it could reduce myocardial infarct size and ischemia/reperfusion injury in both non-diabetic and diabetic conditions. It has been shown that TXL could regulate peroxisome proliferator activated receptor-α (PPAR-α), a positive modulator of angiopoietin-like 4 (Angptl4), in diabetic rats. Endothelial junction substructure components, such as VE-cadherin, are involved in the protection of reperfusion injury. Thus, we hypothesized cell-intrinsic and endothelial-specific Angptl4 mediated the protection of TXL on endothelial barrier under high glucose condition against ischemia/reperfusion-injury via PPAR-α pathway. METHODS Incubated with high glucose medium, the human cardiac microvascular endothelial cells (HCMECs) were then exposed to oxygen-glucose-serum deprivation (2 hours) and restoration (2 hours) stimulation, with or without TXL, insulin, or rhAngptl4 pretreatment. RESULTS TXL, insulin, and rhAngptl4 had similar protective effects on the endothelial barrier. TXL treatment reversed the endothelial barrier breakdown in HCMECs significantly as identified by decreasing endothelial permeability, upregulating the expression of JAM-A, VE-cadherin, and integrin-α5 and increasing the membrane location of VE-cadherin and integrin-α5, and these effects of TXL were as effective as insulin and rhAngptl4. However, Angptl4 knock-down with small interfering RNA (siRNA) interference and PPAR-α inhibitor MK886 partially abrogated these beneficial effects of TXL. Western blotting also revealed that similar with insulin, TXL upregulated the expression of Angptl4 in HCMECs, which could be inhibited by Angptl4 siRNA or MK886 exposure. TXL treatment increased PPAR-α activity, which could be diminished by MK886 but not by Angptl4 siRNA. CONCLUSION These data suggest cell-intrinsic and endothelial-specific Angptl4 mediates the protection of TXL against endothelial barrier breakdown during oxygen-glucose-serum deprivation and restoration under high glucose condition partly via the PPAR-α/Angptl4 pathway.
Collapse
Affiliation(s)
- Kang Qi
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuejin Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongjian Geng
- Department of Internal Medicine, Division of Cardiology, The University of Texas Health Science Center at Houston, Houston, TX
| | - Hehe Cui
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangdong Li
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Jin
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guihao Chen
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaqiu Tian
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianmin Meng
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
15
|
Ding S, Wu D, Lu Q, Qian L, Ding Y, Liu G, Jia X, Zhang Y, Xiao W, Gong W. Angiopoietin-like 4 deficiency upregulates macrophage function through the dysregulation of cell-intrinsic fatty acid metabolism. Am J Cancer Res 2020; 10:595-609. [PMID: 32195030 PMCID: PMC7061760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 01/27/2020] [Indexed: 06/10/2023] Open
Abstract
Angiopoietin-like 4 (ANGPLT4) regulates lipid metabolism by inhibiting lipoprotein lipase. Abnormal ANGTPL4 levels are associated with metabolic syndrome, atherosclerosis, inflammation, and cancer. We show here that ANGPTL4-deficient mice have abnormally large numbers of macrophages in the spleen, and that these macrophages produce large amounts of TNF-α, CD86, and inducible nitric oxide synthase. However, recombinant ANGPTL4 protein did not inhibit macrophage function ex vivo. Glycolysis and fatty-acid synthesis were upregulated in ANGPTL4-/- macrophages, whereas fatty-acid oxidation was decreased. Elevated levels of free fatty acids in the cytoplasm of ANGPTL4-/- macrophages were confirmed. ANGPTL4-/- macrophages also displayed endoplasmic reticulum (ER) stress after stimulation with LPS. Protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling was activated, but no major change in liver kinase B1 (LKB1)/adenosine 5'-monophosphate (AMP)- activated protein kinase (AMPK) phosphorylation was observed in ANGPTL4-/- macrophages. The modulation of fatty-acid metabolism prevented ER stress and the expression of inflammatory molecules, but the activation of ANGPTL4-/- macrophages was not restored by the inhibition of glycolysis. Thus, ANGPTL4 deficiency in macrophages results in ER stress due to the cell-intrinsic reprogramming of fatty-acid metabolism. Intracellular ANGPLT4 expression could thus be manipulated to modulate macrophage function.
Collapse
Affiliation(s)
- Shizhen Ding
- Department of Immunology, School of Medicine, Yangzhou UniversityYangzhou 225001, P. R. China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile DiseasesYangzhou 225001, P. R. China
| | - Dandan Wu
- Department of Immunology, School of Medicine, Yangzhou UniversityYangzhou 225001, P. R. China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile DiseasesYangzhou 225001, P. R. China
| | - Quotao Lu
- Department of Gastroenterology, Affiliated Hospital, Yangzhou UniversityYangzhou 225001, P. R. China
| | - Li Qian
- Department of Immunology, School of Medicine, Yangzhou UniversityYangzhou 225001, P. R. China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile DiseasesYangzhou 225001, P. R. China
| | - Yanbing Ding
- Department of Gastroenterology, Affiliated Hospital, Yangzhou UniversityYangzhou 225001, P. R. China
| | - George Liu
- Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Institute of Cardiovascular Science, Peking UniversityBeijing 100191, P. R. China
| | - Xiaoqin Jia
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile DiseasesYangzhou 225001, P. R. China
- Jiangsu Key Laboratory of ZoonosisYangzhou 225001, P. R. China
| | - Yu Zhang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile DiseasesYangzhou 225001, P. R. China
- Jiangsu Key Laboratory of ZoonosisYangzhou 225001, P. R. China
| | - Weiming Xiao
- Department of Gastroenterology, Affiliated Hospital, Yangzhou UniversityYangzhou 225001, P. R. China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile DiseasesYangzhou 225001, P. R. China
- Jiangsu Key Laboratory of ZoonosisYangzhou 225001, P. R. China
| | - Weijuan Gong
- Department of Immunology, School of Medicine, Yangzhou UniversityYangzhou 225001, P. R. China
- Department of Gastroenterology, Affiliated Hospital, Yangzhou UniversityYangzhou 225001, P. R. China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile DiseasesYangzhou 225001, P. R. China
- Jiangsu Key Laboratory of ZoonosisYangzhou 225001, P. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou 225001, P. R. China
| |
Collapse
|
16
|
Huang H, Kolibabka M, Eshwaran R, Chatterjee A, Schlotterer A, Willer H, Bieback K, Hammes HP, Feng Y. Intravitreal injection of mesenchymal stem cells evokes retinal vascular damage in rats. FASEB J 2019; 33:14668-14679. [DOI: 10.1096/fj.201901500r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Hongpeng Huang
- Experimental Pharmacology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias Kolibabka
- Fifth Medical Clinic, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Rachana Eshwaran
- Experimental Pharmacology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anupriya Chatterjee
- Experimental Pharmacology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andrea Schlotterer
- Fifth Medical Clinic, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hélène Willer
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hans-Peter Hammes
- Fifth Medical Clinic, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yuxi Feng
- Experimental Pharmacology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
17
|
Sodhi A, Ma T, Menon D, Deshpande M, Jee K, Dinabandhu A, Vancel J, Lu D, Montaner S. Angiopoietin-like 4 binds neuropilins and cooperates with VEGF to induce diabetic macular edema. J Clin Invest 2019; 129:4593-4608. [PMID: 31545295 PMCID: PMC6819094 DOI: 10.1172/jci120879] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/01/2019] [Indexed: 02/06/2023] Open
Abstract
The majority of patients with diabetic macular edema (DME), the most common cause of vision loss in working-age Americans, do not respond adequately to current therapies targeting VEGFA. Here, we show that expression of angiopoietin-like 4 (ANGPTL4), a HIF-1-regulated gene product, is increased in the eyes of diabetic mice and patients with DME. We observed that ANGPTL4 and VEGF act synergistically to destabilize the retinal vascular barrier. Interestingly, while ANGPTL4 modestly enhanced tyrosine phosphorylation of VEGF receptor 2, promotion of vascular permeability by ANGPTL4 was independent of this receptor. Instead, we found that ANGPTL4 binds directly to neuropilin 1 (NRP1) and NRP2 on endothelial cells (ECs), leading to rapid activation of the RhoA/ROCK signaling pathway and breakdown of EC-EC junctions. Treatment with a soluble fragment of NRP1 (sNRP1) prevented ANGPTL4 from binding to NRP1 and blocked ANGPTL4-induced activation of RhoA as well as EC permeability in vitro and retinal vascular leakage in diabetic animals in vivo. In addition, sNRP1 reduced the stimulation of EC permeability by aqueous fluid from patients with DME. Collectively, these data identify the ANGPTL4/NRP/RhoA pathway as a therapeutic target for the treatment of DME.
Collapse
Affiliation(s)
- Akrit Sodhi
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Tao Ma
- Department of Oncology and Diagnostic Sciences, School of Dentistry, and
| | - Deepak Menon
- Department of Oncology and Diagnostic Sciences, School of Dentistry, and
| | - Monika Deshpande
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kathleen Jee
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Jordan Vancel
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Daoyuan Lu
- Department of Oncology and Diagnostic Sciences, School of Dentistry, and
| | - Silvia Montaner
- Department of Oncology and Diagnostic Sciences, School of Dentistry, and,Greenebaum Cancer Center, University of Maryland, Baltimore (UMB), Maryland, USA
| |
Collapse
|
18
|
Cossutta M, Darche M, Carpentier G, Houppe C, Ponzo M, Raineri F, Vallée B, Gilles ME, Villain D, Picard E, Casari C, Denis C, Paques M, Courty J, Cascone I. Weibel-Palade Bodies Orchestrate Pericytes During Angiogenesis. Arterioscler Thromb Vasc Biol 2019; 39:1843-1858. [PMID: 31315435 DOI: 10.1161/atvbaha.119.313021] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Objective Weibel-Palade bodies (WPBs) are endothelial cell (EC)-specific organelles formed by vWF (von Willebrand factor) polymerization and that contain the proangiogenic factor Ang-2 (angiopoietin-2). WPB exocytosis has been shown to be implicated for vascular repair and inflammatory responses. Here, we investigate the role of WPBs during angiogenesis and vessel stabilization. Approach and Results WPB density in ECs decreased at the angiogenic front of retinal vascular network during development and neovascularization compared with stable vessels. In vitro, VEGF (vascular endothelial growth factor) induced a VEGFR-2 (vascular endothelial growth factor receptor-2)-dependent exocytosis of WPBs that contain Ang-2 and consequently the secretion of vWF and Ang-2. Blocking VEGF-dependant WPB exocytosis and Ang-2 secretion promoted pericyte migration toward ECs. Pericyte migration was inhibited by adding recombinant Ang-2 or by silencing Ang-1 (angiopoietin-1) or Tie2 (angiopoietin-1 receptor) in pericytes. Consistently, in vivo anti-VEGF treatment induced accumulation of WPBs in retinal vessels because of the inhibition of WPB exocytosis and promoted the increase of pericyte coverage of retinal vessels during angiogenesis. In tumor angiogenesis, depletion of WPBs in vWF knockout tumor-bearing mice promoted an increase of tumor angiogenesis and a decrease of pericyte coverage of tumor vessels. By another approach, normalized tumor vessels had higher WPB density. Conclusions We demonstrate that WPB exocytosis and Ang-2 secretion are regulated during angiogenesis to limit pericyte coverage of remodeling vessels by disrupting Ang-1/Tie2 autocrine signaling in pericytes.
Collapse
Affiliation(s)
- Mélissande Cossutta
- From the CRRET laboratory, CNRS ERL 9215, University of Paris-Est Créteil (UPEC), France (M.C., M.D., G.C., C.H., M.P., F.R., B.V., M.-E.G., D.V., J.C., I.C.)
| | - Marie Darche
- From the CRRET laboratory, CNRS ERL 9215, University of Paris-Est Créteil (UPEC), France (M.C., M.D., G.C., C.H., M.P., F.R., B.V., M.-E.G., D.V., J.C., I.C.)
| | - Gilles Carpentier
- From the CRRET laboratory, CNRS ERL 9215, University of Paris-Est Créteil (UPEC), France (M.C., M.D., G.C., C.H., M.P., F.R., B.V., M.-E.G., D.V., J.C., I.C.)
| | - Claire Houppe
- From the CRRET laboratory, CNRS ERL 9215, University of Paris-Est Créteil (UPEC), France (M.C., M.D., G.C., C.H., M.P., F.R., B.V., M.-E.G., D.V., J.C., I.C.)
| | - Matteo Ponzo
- From the CRRET laboratory, CNRS ERL 9215, University of Paris-Est Créteil (UPEC), France (M.C., M.D., G.C., C.H., M.P., F.R., B.V., M.-E.G., D.V., J.C., I.C.).,Quinze Vingts National Ophthalmology Hospital, Paris, France (M.P.)
| | - Fabio Raineri
- From the CRRET laboratory, CNRS ERL 9215, University of Paris-Est Créteil (UPEC), France (M.C., M.D., G.C., C.H., M.P., F.R., B.V., M.-E.G., D.V., J.C., I.C.)
| | - Benoit Vallée
- From the CRRET laboratory, CNRS ERL 9215, University of Paris-Est Créteil (UPEC), France (M.C., M.D., G.C., C.H., M.P., F.R., B.V., M.-E.G., D.V., J.C., I.C.)
| | - Maud-Emmanuelle Gilles
- From the CRRET laboratory, CNRS ERL 9215, University of Paris-Est Créteil (UPEC), France (M.C., M.D., G.C., C.H., M.P., F.R., B.V., M.-E.G., D.V., J.C., I.C.)
| | - Delphine Villain
- From the CRRET laboratory, CNRS ERL 9215, University of Paris-Est Créteil (UPEC), France (M.C., M.D., G.C., C.H., M.P., F.R., B.V., M.-E.G., D.V., J.C., I.C.)
| | - Emilie Picard
- Inserm, U1138, Team 17, Physiopathology of Ocular Diseases to Clinical Development, University of Paris Descartes Sorbonne Paris Cité, Cordeliers Research Center, France (E.P.)
| | - Caterina Casari
- Inserm, UMR S1176, Paris-Sud University, Paris-Saclay University, Le Kremlin-Bicêtre, France (C.C., C.D.)
| | - Cécile Denis
- Inserm, UMR S1176, Paris-Sud University, Paris-Saclay University, Le Kremlin-Bicêtre, France (C.C., C.D.)
| | - Michel Paques
- Department of Therapeutics, Sorbonne University, INSERM, CNRS, Vision Institute, Paris, France (M.P.)
| | - José Courty
- From the CRRET laboratory, CNRS ERL 9215, University of Paris-Est Créteil (UPEC), France (M.C., M.D., G.C., C.H., M.P., F.R., B.V., M.-E.G., D.V., J.C., I.C.)
| | - Ilaria Cascone
- From the CRRET laboratory, CNRS ERL 9215, University of Paris-Est Créteil (UPEC), France (M.C., M.D., G.C., C.H., M.P., F.R., B.V., M.-E.G., D.V., J.C., I.C.)
| |
Collapse
|
19
|
Budi EH, Mamai O, Hoffman S, Akhurst RJ, Derynck R. Enhanced TGF-β Signaling Contributes to the Insulin-Induced Angiogenic Responses of Endothelial Cells. iScience 2019; 11:474-491. [PMID: 30684493 PMCID: PMC6348203 DOI: 10.1016/j.isci.2018.12.038] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 11/12/2018] [Accepted: 12/28/2018] [Indexed: 12/17/2022] Open
Abstract
Angiogenesis, the development of new blood vessels, is a key process in disease. We reported that insulin promotes translocation of transforming growth factor β (TGF-β) receptors to the plasma membrane of epithelial and fibroblast cells, thus enhancing TGF-β responsiveness. Since insulin promotes angiogenesis, we addressed whether increased autocrine TGF-β signaling participates in endothelial cell responses to insulin. We show that insulin enhances TGF-β responsiveness and autocrine TGF-β signaling in primary human endothelial cells, by inducing a rapid increase in cell surface TGF-β receptor levels. Autocrine TGF-β/Smad signaling contributed substantially to insulin-induced gene expression associated with angiogenesis, including TGF-β target genes encoding angiogenic mediators; was essential for endothelial cell migration; and participated in endothelial cell invasion and network formation. Blocking TGF-β signaling impaired insulin-induced microvessel outgrowth from neonatal aortic rings and modified insulin-stimulated blood vessel formation in zebrafish. We conclude that enhanced autocrine TGF-β signaling is integral to endothelial cell and angiogenic responses to insulin.
Collapse
Affiliation(s)
- Erine H Budi
- Department of Cell and Tissue Biology, University of California at San Francisco Broad Center, Room RMB-1027, 35 Medical Center Way, San Francisco, CA 94143-0669, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Ons Mamai
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Steven Hoffman
- Department of Cell and Tissue Biology, University of California at San Francisco Broad Center, Room RMB-1027, 35 Medical Center Way, San Francisco, CA 94143-0669, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Rosemary J Akhurst
- Department of Anatomy, University of California at San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Rik Derynck
- Department of Cell and Tissue Biology, University of California at San Francisco Broad Center, Room RMB-1027, 35 Medical Center Way, San Francisco, CA 94143-0669, USA; Department of Anatomy, University of California at San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
20
|
Kaushik G, Gil DA, Torr E, Berge ES, Soref C, Uhl P, Fontana G, Antosiewicz-Bourget J, Edington C, Schwartz MP, Griffith LG, Thomson JA, Skala MC, Daly WT, Murphy WL. Quantitative Label-Free Imaging of 3D Vascular Networks Self-Assembled in Synthetic Hydrogels. Adv Healthc Mater 2019; 8:e1801186. [PMID: 30565891 PMCID: PMC6601624 DOI: 10.1002/adhm.201801186] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 11/22/2018] [Indexed: 12/17/2022]
Abstract
Vascularization is an important strategy to overcome diffusion limits and enable the formation of complex, physiologically relevant engineered tissues and organoids. Self-assembly is a technique to generate in vitro vascular networks, but engineering the necessary network morphology and function remains challenging. Here, autofluorescence multiphoton microscopy (aMPM), a label-free imaging technique, is used to quantitatively evaluate in vitro vascular network morphology. Vascular networks are generated using human embryonic stem cell-derived endothelial cells and primary human pericytes encapsulated in synthetic poly(ethylene glycol)-based hydrogels. Two custom-built bioreactors are used to generate distinct fluid flow patterns during vascular network formation: recirculating flow or continuous flow. aMPM is used to image these 3D vascular networks without the need for fixation, labels, or dyes. Image processing and analysis algorithms are developed to extract quantitative morphological parameters from these label-free images. It is observed with aMPM that both bioreactors promote formation of vascular networks with lower network anisotropy compared to static conditions, and the continuous flow bioreactor induces more branch points compared to static conditions. Importantly, these results agree with trends observed with immunocytochemistry. These studies demonstrate that aMPM allows label-free monitoring of vascular network morphology to streamline optimization of growth conditions and provide quality control of engineered tissues.
Collapse
Affiliation(s)
- Gaurav Kaushik
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
- Human Models for Analysis of Pathways (HMAPs) Center, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
| | - Daniel A Gil
- Morgridge Institute for Research, 330 North Orchard Street, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| | - Elizabeth Torr
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
- Human Models for Analysis of Pathways (HMAPs) Center, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
| | - Elizabeth S Berge
- Morgridge Institute for Research, 330 North Orchard Street, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| | - Cheryl Soref
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
- Human Models for Analysis of Pathways (HMAPs) Center, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
| | - Peyton Uhl
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
- Human Models for Analysis of Pathways (HMAPs) Center, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
| | - Gianluca Fontana
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
- Human Models for Analysis of Pathways (HMAPs) Center, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
| | - Jessica Antosiewicz-Bourget
- Human Models for Analysis of Pathways (HMAPs) Center, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
- Morgridge Institute for Research, 330 North Orchard Street, Madison, WI, 53715, USA
| | - Collin Edington
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Michael P Schwartz
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - James A Thomson
- Human Models for Analysis of Pathways (HMAPs) Center, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
- Morgridge Institute for Research, 330 North Orchard Street, Madison, WI, 53715, USA
| | - Melissa C Skala
- Human Models for Analysis of Pathways (HMAPs) Center, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
- Morgridge Institute for Research, 330 North Orchard Street, Madison, WI, 53715, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - William T Daly
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
- Human Models for Analysis of Pathways (HMAPs) Center, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
| | - William L Murphy
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
- Human Models for Analysis of Pathways (HMAPs) Center, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5418, Madison, WI, 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| |
Collapse
|
21
|
Yang X, Cheng Y, Su G. A review of the multifunctionality of angiopoietin-like 4 in eye disease. Biosci Rep 2018; 38:BSR20180557. [PMID: 30049845 PMCID: PMC6137252 DOI: 10.1042/bsr20180557] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/02/2018] [Accepted: 07/18/2018] [Indexed: 12/12/2022] Open
Abstract
Angiopoietin-like protein 4 (ANGPTL4) is a multifunctional cytokine regulating vascular permeability, angiogenesis, and inflammation. Dysregulations in these responses contribute to the pathogenesis of ischemic retinopathies such as diabetic retinopathy (DR), age-related macular degeneration (AMD), retinal vein occlusion, and sickle cell retinopathy (SCR). However, the role of ANGPTL4 in these diseases remains controversial. Here, we summarize the functional mechanisms of ANGPTL4 in several diseases. We highlight original studies that provide detailed data about the mechanisms of action for ANGPTL4, its applications as a diagnostic or prognostic biomarker, and its use as a potential therapeutic target. Taken together, the discussions in this review will help us gain a better understanding of the molecular mechanisms by which ANGPTL4 functions in eye disease and will provide directions for future research.
Collapse
Affiliation(s)
- Xinyue Yang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Yan Cheng
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Guanfang Su
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130041, China
| |
Collapse
|
22
|
Marchand M, Monnot C, Muller L, Germain S. Extracellular matrix scaffolding in angiogenesis and capillary homeostasis. Semin Cell Dev Biol 2018; 89:147-156. [PMID: 30165150 DOI: 10.1016/j.semcdb.2018.08.007] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/31/2018] [Accepted: 08/14/2018] [Indexed: 01/03/2023]
Abstract
The extracellular matrix (ECM) of blood vessels, which is composed of both the vascular basement membrane (BM) and the interstitial ECM is identified as a crucial component of the vasculature. We here focus on the unique molecular composition and scaffolding of the capillary ECM, which provides structural support to blood vessels and regulates properties of endothelial cells and pericytes. The major components of the BM are collagen IV, laminins, heparan sulfate proteoglycans and nidogen and also associated proteins such as collagen XVIII and fibronectin. Their organization and scaffolding in the BM is required for proper capillary morphogenesis and maintenance of vascular homeostasis. The BM also regulates vascular mechanosensing. A better understanding of the mechanical and structural properties of the vascular BM and interstitial ECM therefore opens new perspectives to control physiological and pathological angiogenesis and vascular homeostasis. The overall aim of this review is to explain how ECM scaffolding influences angiogenesis and capillary integrity.
Collapse
Affiliation(s)
- Marion Marchand
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 11 Place Marcelin Berthelot, 75005, Paris, France; Sorbonne Université, Collège Doctoral, F-75005 Paris, France
| | - Catherine Monnot
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 11 Place Marcelin Berthelot, 75005, Paris, France
| | - Laurent Muller
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 11 Place Marcelin Berthelot, 75005, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 11 Place Marcelin Berthelot, 75005, Paris, France.
| |
Collapse
|
23
|
Rienks M, Carai P, van Teeffelen J, Eskens B, Verhesen W, Hemmeryckx B, Johnson DM, van Leeuwen R, Jones EA, Heymans S, Papageorgiou AP. SPARC preserves endothelial glycocalyx integrity, and protects against adverse cardiac inflammation and injury during viral myocarditis. Matrix Biol 2018; 74:21-34. [PMID: 29730504 DOI: 10.1016/j.matbio.2018.04.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 12/24/2022]
Abstract
Myocardial damage as a consequence of cardiotropic viruses leads to a broad variety of clinical presentations and is still a complicated condition to diagnose and treat. Whereas the extracellular matrix protein Secreted Protein Acidic and Rich in Cysteine or SPARC has been implicated in hypertensive and ischemic heart disease by modulating collagen production and cross-linking, its role in cardiac inflammation and endothelial function is yet unknown. Absence of SPARC in mice resulted in increased cardiac inflammation and mortality, and reduced cardiac systolic function upon coxsackievirus-B3 induced myocarditis. Intra-vital microscopic imaging of the microvasculature of the cremaster muscle combined with electron microscopic imaging of the microvasculature of the cardiac muscle uncovered the significance of SPARC in maintaining endothelial glycocalyx integrity and subsequent barrier properties to stop inflammation. Moreover, systemic administration of recombinant SPARC restored the endothelial glycocalyx and consequently reversed the increase in inflammation and mortality observed in SPARC KO mice in response to viral exposure. Reducing the glycocalyx in vivo by systemic administration of hyaluronidase, an enzyme that degrades the endothelial glycocalyx, mimicked the barrier defects found in SPARC KO mice, which could be restored by subsequent administration of recombinant SPARC. In conclusion, the secreted glycoprotein SPARC protects against adverse cardiac inflammation and mortality by improving the glycocalyx function and resulting endothelial barrier function during viral myocarditis.
Collapse
Affiliation(s)
- Marieke Rienks
- Cardiovascular Department, King's College London, United Kingdom; Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, The Netherlands.
| | - Paolo Carai
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, The Netherlands
| | | | - Bart Eskens
- Department of Physiology, Maastricht University, The Netherlands
| | - Wouter Verhesen
- Cardiovascular Department, King's College London, United Kingdom
| | - Bianca Hemmeryckx
- Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU, Leuven, Belgium
| | - Daniel M Johnson
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, The Netherlands
| | - Rick van Leeuwen
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, The Netherlands
| | - Elizabeth A Jones
- Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU, Leuven, Belgium
| | - Stephane Heymans
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, The Netherlands; Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU, Leuven, Belgium; Netherlands Heart Institute, ICIN, Utrecht, The Netherlands
| | - Anna-Pia Papageorgiou
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, The Netherlands; Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU, Leuven, Belgium
| |
Collapse
|
24
|
Angiopoietin-Like Proteins in Angiogenesis, Inflammation and Cancer. Int J Mol Sci 2018; 19:ijms19020431. [PMID: 29389861 PMCID: PMC5855653 DOI: 10.3390/ijms19020431] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 12/27/2022] Open
Abstract
Altered expression of secreted factors by tumor cells or cells of the tumor microenvironment is a key event in cancer development and progression. In the last decade, emerging evidences supported the autocrine and paracrine activity of the members of the Angiopoietin-like (ANGPTL) protein family in angiogenesis, inflammation and in the regulation of different steps of carcinogenesis and metastasis development. Thus, ANGPTL proteins become attractive either as prognostic or predictive biomarkers, or as novel target for cancer treatment. Here, we outline the current knowledge about the functions of the ANGPTL proteins in angiogenesis, cancer progression and metastasis. Moreover, we discuss the most recent evidences sustaining their role as prognostic or predictive biomarkers for cancer therapy. Although the role of ANGPTL proteins in cancer has not been fully elucidated, increasing evidence suggest their key effects in the proliferative and invasive properties of cancer cells. Moreover, given the common overexpression of ANGPTL proteins in several aggressive solid tumors, and their role in tumor cells and cells of the tumor microenvironment, the field of research about ANGPTL proteins network may highlight new potential targets for the development of future therapeutic strategies.
Collapse
|
25
|
Changes in nitric oxide, angiotensin II, angiopoietin-like protein 4 mRNA, neuregulin 1 mRNA, and platelet endothelial cell adhesion molecule-1 in rats with acute blood stasis induced by high-molecular-weight dextran. J TRADIT CHIN MED 2017. [DOI: 10.1016/s0254-6272(18)30050-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
26
|
Angiopoietin-like 4 Is a Wnt Signaling Antagonist that Promotes LRP6 Turnover. Dev Cell 2017; 43:71-82.e6. [PMID: 29017031 DOI: 10.1016/j.devcel.2017.09.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 08/25/2017] [Accepted: 09/13/2017] [Indexed: 11/23/2022]
Abstract
Angiopoietin-like 4 (ANGPTL4) is a secreted signaling protein that is implicated in cardiovascular disease, metabolic disorder, and cancer. Outside of its role in lipid metabolism, ANGPTL4 signaling remains poorly understood. Here, we identify ANGPTL4 as a Wnt signaling antagonist that binds to syndecans and forms a ternary complex with the Wnt co-receptor Lipoprotein receptor-related protein 6 (LRP6). This protein complex is internalized via clathrin-mediated endocytosis and degraded in lysosomes, leading to attenuation of Wnt/β-catenin signaling. Angptl4 is expressed in the Spemann organizer of Xenopus embryos and acts as a Wnt antagonist to promote notochord formation and prevent muscle differentiation. This unexpected function of ANGPTL4 invites re-interpretation of its diverse physiological effects in light of Wnt signaling and may open therapeutic avenues for human disease.
Collapse
|
27
|
ANGPTL-4 induces diabetic retinal inflammation by activating Profilin-1. Exp Eye Res 2017; 166:140-150. [PMID: 29031854 DOI: 10.1016/j.exer.2017.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/10/2017] [Accepted: 10/10/2017] [Indexed: 12/30/2022]
Abstract
Diabetic retinopathy (DR), the most common cause of irreversible blindness in working-age adults, results in central vision loss that is caused by microvascular damage to the inner lining of the back of the eye, the retina. The aim of this work was to assess the temporal relationships between angiopoietin-like protein-4 (ANGPTL-4), a novel adipocytokine factor, and diabetic retinal inflammation and microvascular dysfunction. The downstream pathway(s) and upstream mediator(s) of ANGPTL-4 were then determined under high glucose (HG) conditions. Diabetic rats and control animals were randomly assigned to receive hypoxia inducible factor-1 alpha (HIF-1α) blockade (doxorubicin or shRNA) or vehicle for 8 weeks. Human retinal microvascular endothelial cells (HRMECs) were incubated with normal or high glucose, with or without blockade or recombinant proteins, for ANGPTL-4, HIF-1α, and vascular endothelial growth factor (VEGF). The levels of ANGPTL-4, profilin-1, HIF-1α, VEGF, interleukin 1 beta (IL-1β), IL-6, and intercellular adherent molecule 1 (ICAM-1) in the rat retinas and HRMEC extracts were examined by Western blotting and real-time RT-PCR. The levels of ANGPTL-4, profilin-1, HIF-1α, and VEGF protein and mRNA were significantly higher in the diabetic rats and HG-exposed HRMECs. ANGPTL-4 was a potent modulator of increased inflammation, permeability, and angiogenesis via activation of the profilin-1 signaling pathway. Our results showed that ANGPTL-4 upregulation was induced by HG, which was dependent on HIF-1α activation that was also triggered by HG, both in vivo and in vitro. Our results suggest that targeting ANGPTL-4, alone or in combination with profilin-1, may be an effective therapeutic strategy and diagnostic screening biomarker for proliferative diabetic retinopathy and other vitreous-retinal inflammatory diseases.
Collapse
|
28
|
Meng Q, Qin Y, Deshpande M, Kashiwabuchi F, Rodrigues M, Lu Q, Ren H, Elisseeff JH, Semenza GL, Montaner SV, Sodhi A. Hypoxia-Inducible Factor-Dependent Expression of Angiopoietin-Like 4 by Conjunctival Epithelial Cells Promotes the Angiogenic Phenotype of Pterygia. Invest Ophthalmol Vis Sci 2017; 58:4514-4523. [PMID: 28873177 PMCID: PMC5584708 DOI: 10.1167/iovs.17-21974] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Purpose Disappointing results from clinical studies assessing the efficacy of therapies targeting vascular endothelial growth factor (VEGF) for the treatment of pterygia suggest that other angiogenic mediators may also play a role in its development. We therefore explore the relative contribution of VEGF, hypoxia-inducible factor (HIF)-1α (the transcription factor that regulates VEGF expression in ocular neovascular disease), and a second HIF-regulated mediator, angiopoietin-like 4 (ANGPTL4), to the angiogenic phenotype of pterygia. Methods Expression of HIF-1α, VEGF, and ANGPTL4 were examined in surgically excised pterygia, and in immortalized human (ih) and primary rabbit (pr) conjunctival epithelial cells (CjECs). Endothelial cell (EC) tubule formation assays using media conditioned by ihCjECs in the presence or absence of inducers/inhibitors of HIF-1 or RNA interference (RNAi) targeting VEGF, ANGPTL4, or both were used to assess their relative contribution to the angiogenic potential of these cells. Results HIF-1α and VEGF expression were detected in 6/6 surgically excised pterygia and localized to CjECs. Accumulation of HIF-1α in was confirmed in ihCjECs and prCjECs, including stratified prCjECs grown on collagen vitrigel, and resulted in expression of VEGF and the promotion of EC tubule formation; the latter effect was partially blocked using RNAi targeting VEGF mRNA expression. We demonstrate expression of a second HIF-regulated angiogenic mediator, ANGPTL4, in CjECs in culture and in surgically excised pterygia. RNAi targeting ANGPTL4 inhibited EC tubule formation and was additive to RNAi targeting VEGF. Conclusions Our results support the development of therapies targeting both ANGPTL4 and VEGF for the treatment of patients with pterygia.
Collapse
Affiliation(s)
- Qianli Meng
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,Guangdong Eye Institute, Department of Ophthalmology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou Guangdong, China
| | - Yaowu Qin
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,EENT Hospital, Fudan University, Shanghai, China
| | - Monika Deshpande
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Fabiana Kashiwabuchi
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Murilo Rodrigues
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Qiaozhi Lu
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Hui Ren
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,EENT Hospital, Fudan University, Shanghai, China
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Gregg L. Semenza
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,Department of Radiation Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Silvia V. Montaner
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland, United States
,Department of Pathology, School of Medicine, University of Maryland, Baltimore, Maryland, United States
,Greenebaum Cancer Center, University of Maryland, Baltimore, Maryland, United States
| | - Akrit Sodhi
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
29
|
Retinal and choroidal angiogenesis: a review of new targets. Int J Retina Vitreous 2017; 3:31. [PMID: 28835854 PMCID: PMC5563895 DOI: 10.1186/s40942-017-0084-9] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/01/2017] [Indexed: 11/10/2022] Open
Abstract
Retinal and choroidal neovascularization are a major cause of significant visual impairment, worldwide. Understanding the various factors involved in the accompanying physiopathology is vital for development of novel treatments, and most important, for preserving patient vision. The intraocular use of anti-vascular endothelial growth factor therapeutics has improved management of the retinal and choroidal neovascularization but some patients do not respond, suggesting other vascular mediators may also contribute to ocular angiogenesis. Several recent studies examined possible new targets for future anti-angiogenic therapies. Potential targets of retinal and choroidal neovascularization therapy include members of the platelet-derived growth factor family, vascular endothelial growth factor sub-family, epidermal growth factor family, fibroblast growth factor family, transforming growth factor-β superfamily (TGF-β1, activins, follistatin and bone morphogenetic proteins), angiopoietin-like family, galectins family, integrin superfamily, as well as pigment epithelium derived factor, hepatocyte growth factor, angiopoietins, endothelins, hypoxia-inducible factors, insulin-like growth factors, cytokines, matrix metalloproteinases and their inhibitors and glycosylation proteins. This review highlights current antiangiogenic therapies under development, and discusses future retinal and choroidal pro- and anti-angiogenic targets as wells as the importance of developing of new drugs.
Collapse
|
30
|
Howell MD, Ottesen EW, Singh NN, Anderson RL, Seo J, Sivanesan S, Whitley EM, Singh RN. TIA1 is a gender-specific disease modifier of a mild mouse model of spinal muscular atrophy. Sci Rep 2017; 7:7183. [PMID: 28775379 PMCID: PMC5543135 DOI: 10.1038/s41598-017-07468-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/26/2017] [Indexed: 02/08/2023] Open
Abstract
Spinal muscular atrophy (SMA) is caused by deletions or mutations of Survival Motor Neuron 1 (SMN1) gene. The nearly identical SMN2 cannot compensate for SMN1 loss due to exon 7 skipping. The allele C (C +/+) mouse recapitulates a mild SMA-like phenotype and offers an ideal system to monitor the role of disease-modifying factors over a long time. T-cell-restricted intracellular antigen 1 (TIA1) regulates SMN exon 7 splicing. TIA1 is reported to be downregulated in obese patients, although it is not known if the effect is gender-specific. We show that female Tia1-knockout (Tia1 -/-) mice gain significant body weight (BW) during early postnatal development. We next examined the effect of Tia1 deletion in novel C +/+/Tia1 -/- mice. Underscoring the opposing effects of Tia1 deletion and low SMN level on BW gain, both C +/+ and C +/+/Tia1 -/- females showed similar BW gain trajectory at all time points during our study. We observed early tail necrosis in C +/+/Tia1 -/- females but not in males. We show enhanced impairment of male reproductive organ development and exacerbation of the C +/+/Tia1 -/- testis transcriptome. Our findings implicate a protein factor as a gender-specific modifier of a mild mouse model of SMA.
Collapse
Affiliation(s)
- Matthew D Howell
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Eric W Ottesen
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Natalia N Singh
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Rachel L Anderson
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Joonbae Seo
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | | | - Elizabeth M Whitley
- Department of Veterinary Pathology, Iowa State University, Ames, IA, 50011-1250, USA
- Pathogenesis, LLC, Gainesville, Florida, 32614, USA
| | - Ravindra N Singh
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA.
| |
Collapse
|
31
|
Li B, Qian M, Cao H, Jia Q, Wu Z, Yang X, Ma T, Wei H, Chen T, Xiao J. TGF-β2-induced ANGPTL4 expression promotes tumor progression and osteoclast differentiation in giant cell tumor of bone. Oncotarget 2017; 8:54966-54977. [PMID: 28903395 PMCID: PMC5589634 DOI: 10.18632/oncotarget.18629] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/29/2017] [Indexed: 11/25/2022] Open
Abstract
Although emerging studies have implicated that Aiopoietin-like 4 Protein (ANGPTL4) is related to the aggressiveness and metastasis of many tumors, the role of ANGPLT4 in giant cell tumor (GCT) of bone was rarely investigated. The mechanism of ANGPLT4 in tumor-induced osteoclastogenesis still remains unclear. In this study, we first demonstrated that ANGPTL4 was highly expressed in GCT compared to normal tissues, while we showed that TGF-β2 released by osteoclasts induced bone resorption could increase the expression of ANGPTL4 in GCTSCs. By using the luciferase reporter assay, we found that two downstreams of TGF-β2, Smad3 and Smad4, could directly activate the promoter of ANGPTL4, which might explain the mechanism of TGF-β2-induced ANGPLT4 expression. Moreover, knockout of ANGPTL4 by TALENs in GCTSCs inhibited tumor growth, angiogenesis and osteoclastogenesis in GCT in vitro. By using the chick chorio-allantoic membrane (CAM) models, we further showed that inhibition of ANGPTL4 suppressed tumor growth and giant cell formation in vivo. In addition, some new pathways involved in ANGPTL4 application were identified through microarray assay, which may partly explain the mechanism of ANGPTL4 in GCT. Taken together, our study for the first time identified the role of ANGPLT4 in GCT of bone, which may provide a new target for the diagnosis and treatment of GCT.
Collapse
Affiliation(s)
- Bo Li
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ming Qian
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hao Cao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Qi Jia
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhipeng Wu
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xinghai Yang
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Tianyi Ma
- Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, China
| | - Haifeng Wei
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Tianrui Chen
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jianru Xiao
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
32
|
Knowles HJ. Multiple Roles of Angiopoietin-Like 4 in Osteolytic Disease. Front Endocrinol (Lausanne) 2017; 8:80. [PMID: 28458654 PMCID: PMC5394121 DOI: 10.3389/fendo.2017.00080] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 03/30/2017] [Indexed: 12/17/2022] Open
Abstract
Hypoxia and the hypoxia-inducible factor (HIF) transcription factor drive pathological bone loss in conditions including rheumatoid arthritis (RA), osteoarthritis, osteoporosis, primary bone tumours, and bone metastatic cancer. There is therefore considerable interest in determining the function(s) of HIF-induced genes in these pathologies. Angiopoietin-like 4 (ANGPTL4) is an adipose-derived, HIF-1α- and PPARγ-induced gene that was originally discovered as an endocrine and autocrine/paracrine regulator of lipid metabolism. Given the inverse relationship between bone adiposity and fracture risk, ANGPTL4 might be considered a good candidate for mediating the downstream effects of HIF-1α relevant to osteolytic disease. This review will consider the possible roles of ANGPTL4 in regulation of osteoclast-mediated bone resorption, cartilage degradation, angiogenesis, and inflammation, focusing on results obtained in the study of RA. Possible roles in other musculoskeletal pathologies will also be discussed. This will highlight ANGPTL4 as a regulator of multiple disease processes, which could represent a novel therapeutic target in osteolytic musculoskeletal disease.
Collapse
Affiliation(s)
- Helen J. Knowles
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- *Correspondence: Helen J. Knowles,
| |
Collapse
|
33
|
Gomez Perdiguero E, Liabotis-Fontugne A, Durand M, Faye C, Ricard-Blum S, Simonutti M, Augustin S, Robb BM, Paques M, Valenzuela DM, Murphy AJ, Yancopoulos GD, Thurston G, Galaup A, Monnot C, Germain S. ANGPTL4-αvβ3 interaction counteracts hypoxia-induced vascular permeability by modulating Src signalling downstream of vascular endothelial growth factor receptor 2. J Pathol 2016; 240:461-471. [PMID: 27577973 DOI: 10.1002/path.4805] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 08/22/2016] [Accepted: 08/26/2016] [Indexed: 01/13/2023]
Abstract
Dynamic control of endothelial cell junctions is essential for vascular homeostasis and angiogenesis. We recently provided genetic evidence that ANGPTL4 is a key regulator of vascular integrity both during developmental and in hypoxia-induced pathological conditions. The purpose of the present study was to decipher the molecular mechanisms through which ANGPTL4 regulates vascular integrity. Using surface plasmon resonance and proximity ligation assays, we show that ANGPTL4 binds integrin αvβ3. In vitro and in vivo functional assays with Angptl4-deficient mice demonstrate that ANGPTL4-αvβ3 interaction is necessary to mediate ANGPTL4 vasoprotective effects. Mechanistically, ANGPTL4-αvβ3 interaction enhances Src recruitment to integrin αvβ3 and inhibits Src signalling downstream of vascular endothelial growth factor receptor 2 (VEFGR2), thereby repressing hypoxia-induced breakdown of VEGFR2-VE-cadherin and VEGFR2-αvβ3 complexes. We further demonstrate that intravitreal injection of recombinant human ANGPTL4 limits vascular permeability and leads to increased adherens junction and tight junction integrity. These findings identify a novel mechanism by which ANGPTL4 counteracts hypoxia-driven vascular permeability through integrin αvβ3 binding, modulation of VEGFR2-Src kinase signalling, and endothelial junction stabilization. We further demonstrate that Angptl4-deficient mice show increased vascular leakage in vivo in a model of laser-induced choroidal neovascularization, indicating that this newly identified ANGPTL4-αvβ3 axis might be a target for pharmaceutical intervention in pathological conditions. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Elisa Gomez Perdiguero
- Centre for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France.,Ecole Doctorale 394: Physiologie, Physiopathologie et Thérapeutique, Université Pierre et Marie Curie, Paris, France
| | - Athanasia Liabotis-Fontugne
- Centre for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France.,Ecole Doctorale 394: Physiologie, Physiopathologie et Thérapeutique, Université Pierre et Marie Curie, Paris, France
| | - Mélanie Durand
- Centre for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Clément Faye
- UMR 5086 CNRS Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Sylvie Ricard-Blum
- UMR 5086 CNRS Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Manuel Simonutti
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Sébastien Augustin
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Bryan M Robb
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Michel Paques
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, CNRS, Institut de la Vision, Paris, France.,Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France
| | | | | | | | | | - Ariane Galaup
- Centre for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Catherine Monnot
- Centre for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Stéphane Germain
- Centre for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| |
Collapse
|
34
|
Im JE, Song SH, Suh W. Src tyrosine kinase regulates the stem cell factor-induced breakdown of the blood-retinal barrier. Mol Vis 2016; 22:1213-1220. [PMID: 27746675 PMCID: PMC5063088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 10/11/2016] [Indexed: 10/31/2022] Open
Abstract
PURPOSE Stem cell factor (SCF) has been recently acknowledged as a novel endothelial permeability factor. However, the mechanisms by which SCF-induced activation of the SCF cognate receptor, cKit, enhances endothelial permeability have not been fully elucidated. This study aimed to investigate the role of Src in SCF-induced breakdown of the blood-retinal barrier (BRB). METHODS In vitro endothelial permeability and in vivo retinal vascular permeability assays were performed to investigate the role of Src in SCF-induced breakdown of the BRB. Immunofluorescence staining experiments were performed to analyze the cellular distribution of phosphorylated Src and vascular endothelial (VE)-cadherin. RESULTS SCF markedly reduced electric resistance across the human retinal vascular endothelial monolayer in vitro and enhanced extravasation of dyes in murine retinal vasculature in vivo. Inhibition of cKit activation using cKit mutant mice and chemical inhibitor substantially diminished the ability of SCF to increase endothelial permeability and retinal vascular leakage. In human retinal vascular endothelial cells, SCF induced strong phosphorylation of Src and distinct localization of phosphorylated Src in the plasma membrane. Inhibition of Src activation using chemical inhibitors abolished the SCF-induced hyperpermeability of human retinal vascular endothelial cells and retinal vascular leakage in mice. In addition, treatment with Src inhibitors restored junctional expression of VE-cadherin that disappeared in SCF-treated retinal endothelial cells and retinal vasculature. CONCLUSIONS These results showed the important role of Src in mediating SCF-induced breakdown of the BRB and retinal vascular leakage. Given that increased retinal vascular permeability is a common manifestation of various ocular diseases, the SCF/cKit/Src signaling pathway may be involved in the development of the hyperpermeable retinal vasculature in many ocular disorders.
Collapse
|
35
|
Kremer PHC, Koeleman BPC, Rinkel GJ, Diekstra FP, van den Berg LH, Veldink JH, Klijn CJM. Susceptibility loci for sporadic brain arteriovenous malformation; a replication study and meta-analysis. J Neurol Neurosurg Psychiatry 2016; 87:693-6. [PMID: 26272027 DOI: 10.1136/jnnp-2014-310094] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 07/28/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Case-control studies have reported multiple genetic loci to be associated with sporadic brain arteriovenous malformations (AVMs) but most of these have not been replicated in independent populations. The aim of this study was to find additional evidence for these reported associations and perform a meta-analysis including all previously published results. METHODS We included 167 Dutch patients and 1038 Dutch controls. Case genotyping was performed by KASPar assays. Controls had been previously genotyped with a genome wide single nucleotide polymorphisms (SNP) array. Differences in genotype frequencies between cases and controls were estimated by χ(2) testing in Plink V.1.07. Meta-analysis was performed in RevMan V.5.3. RESULTS In our case-control study we found no significant association with brain AVM (BAVM) for previously discovered SNPs near ANGPTL4, IL-1β, GPR124, VEGFA and MMP-3. The meta-analysis revealed a statistically significant association with BAVMs for the polymorphism rs11672433 near ANGPTL4 (OR 1.39; 95% CI 1.10 to 1.75, p value 0.005). CONCLUSIONS The results of this study support a role for the previously identified SNP near ANGPTL4 in the pathogenesis of AVMs. Previously found associations with SNPs near IL-1β, GPR124, VEGFA and MMP-3 genes could not be substantiated in our replication cohort or in the meta-analysis.
Collapse
Affiliation(s)
- P H C Kremer
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - B P C Koeleman
- Department of Biomedical Genetics and Complex Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - G Je Rinkel
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F P Diekstra
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - L H van den Berg
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J H Veldink
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - C J M Klijn
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
36
|
Liu R, Liu Q, Wang K, Dang X, Zhang F. Comparative analysis of gene expression profiles in normal hip human cartilage and cartilage from patients with necrosis of the femoral head. Arthritis Res Ther 2016; 18:98. [PMID: 27146865 PMCID: PMC4857375 DOI: 10.1186/s13075-016-0991-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 04/07/2016] [Indexed: 12/02/2022] Open
Abstract
Background The pathogenesis of necrosis of the femoral head (NFH) remains elusive. Limited studies were conducted to investigate the molecular mechanism of hip articular cartilage damage in NFH. We conducted genome-wide gene expression profiling of hip articular cartilage with NFH. Methods Hip articular cartilage specimens were collected from 18 NFH patients and 18 healthy controls. Gene expression profiling of NFH articular cartilage was carried out by Agilent Human 4x44K Gene Expression Microarray chip. Differently expressed genes were identified using the significance analysis of microarrays (SAM) software. Gene Ontology (GO) enrichment analysis of differently expressed genes was performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID). Significantly differently expressed genes in the microarray experiment were selected for quantitative real-time PCR (qRT-PCR) and immunohistochemical validation. Results SAM identified 27 differently expressed genes in NFH articular cartilage, functionally involved in extracellular matrix, cytokines, growth factors, cell cycle and apoptosis. The expression patterns of the nine validation genes in qRT-PCR were consistent with that in proteinaceous extracellular matrix (false discovery rate (FDR) = 3.22 × 10-5), extracellular matrix (FDR = 5.78 × 10-5), extracellular region part (FDR = 1.28 × 10-4), collagen (FDR = 3.22 × 10-4), extracellular region (FDR = 4.78 × 10-4) and platelet-derived growth factor binding (FDR = 5.23 × 10-4). Conclusions This study identified a set of differently expressed genes, implicated in articular cartilage damage in NFH. Our study results may provide novel insight into the pathogenesis and rationale of therapies for NFH. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-0991-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ruiyu Liu
- Department of Orthopedics, the Second Affiliated Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China.
| | - Qi Liu
- Department of Orthopedics, the Second Affiliated Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Kunzheng Wang
- Department of Orthopedics, the Second Affiliated Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Xiaoqian Dang
- Department of Orthopedics, the Second Affiliated Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China.
| |
Collapse
|
37
|
Jennewein M, Bubel M, Guthörl S, Metzger W, Weigert M, Pohlemann T, Oberringer M. Two- and three-dimensional co-culture models of soft tissue healing: pericyte-endothelial cell interaction. Cell Tissue Res 2016; 365:279-93. [DOI: 10.1007/s00441-016-2391-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/29/2016] [Indexed: 12/20/2022]
|
38
|
Lu Q, Zou W, Chen B, Zou C, Zhao M, Zheng Z. ANGPTL-4 correlates with vascular endothelial growth factor in patients with proliferative diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol 2015; 254:1281-8. [PMID: 26483143 DOI: 10.1007/s00417-015-3187-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 09/16/2015] [Accepted: 09/30/2015] [Indexed: 01/24/2023] Open
Abstract
PURPOSE To investigate the relationship between angiopoietin-like protein 4 (ANGPTL-4) and vascular endothelial growth factor (VEGF) in the serum and vitreous of eyes in patients with proliferative diabetic retinopathy (PDR). METHODS Thirty-five eyes of 35 patients with PDR, 20 eyes of 20 patients with non-proliferative diabetic retinopathy, 20 eyes of 20 patients with diabetes but no diabetic retinopathy, and 14 eyes of 14 nondiabetic patients with an idiopathic macular hole (IMH) were recruited from Shanghai First People's Hospital. The ANGPTL-4 and VEGF concentrations were determined using enzyme-linked immunosorbent assays. Group means were compared using one-way analysis of variance with GraphPad Prism 4.0 and SPSS ver. 17.0. The research followed the tenets of the Declaration of Helsinki. RESULTS The ANGPTL-4 and VEGF levels were significantly higher in the vitreous and serum of patients with PDR compared with patients with IMH. There were significant correlations between the ANGPTL-4 and VEGF levels in the vitreous and serum of patients with PDR. The vitreous and serum ANGPTL-4 levels were also significantly correlated in patients with PDR. The ANGPTL-4 in both the vitreous and serum correlated with the serum triglyceride and high-density lipoprotein cholesterol levels. CONCLUSIONS The ANGPTL-4 levels were markedly elevated and the ANGPTL-4 expression was directly correlated with the VEGF expression in the vitreous and serum of patients with PDR. The vitreous and serum ANGPTL-4 levels were also significantly correlated with serum lipids in patients with PDR. Our results suggest that the ANGPTL-4 may be used as a new therapeutic target for the treatment of PDR.
Collapse
Affiliation(s)
- Qianyi Lu
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 100 Haining Road, Shanghai, 200080, China
| | - Wenjun Zou
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, No. 68 Zhongshan Road, Wuxi, 214002, China
| | - Bin Chen
- Department of Ophthalmology, Changshu No. 1 People's Hospital, Changshu, 215500, China
| | - Chen Zou
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 100 Haining Road, Shanghai, 200080, China
| | - Minjie Zhao
- Department of Ophthalmology, Nanjing Medical University Affiliated Shanghai General Hospital, Shanghai, 200080, China
| | - Zhi Zheng
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 100 Haining Road, Shanghai, 200080, China.
| |
Collapse
|
39
|
Sun Y, Ju M, Lin Z, Fredrick TW, Evans LP, Tian KT, Saba NJ, Morss PC, Pu WT, Chen J, Stahl A, Joyal JS, Smith LEH. SOCS3 in retinal neurons and glial cells suppresses VEGF signaling to prevent pathological neovascular growth. Sci Signal 2015; 8:ra94. [PMID: 26396267 DOI: 10.1126/scisignal.aaa8695] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neurons and glial cells in the retina contribute to neovascularization, or the formation of abnormal new blood vessels, in proliferative retinopathy, a condition that can lead to vision loss or blindness. We identified a mechanism by which suppressor of cytokine signaling 3 (SOCS3) in neurons and glial cells prevents neovascularization. We found that Socs3 expression was increased in the retinal ganglion cell and inner nuclear layers after oxygen-induced retinopathy. Mice with Socs3 deficiency in neuronal and glial cells had substantially reduced vaso-obliterated retinal areas and increased pathological retinal neovascularization in response to oxygen-induced retinopathy, suggesting that loss of neuronal/glial SOCS3 increased both retinal vascular regrowth and pathological neovascularization. Furthermore, retinal expression of Vegfa (which encodes vascular endothelial growth factor A) was higher in these mice than in Socs3 flox/flox controls, indicating that neuronal and glial SOCS3 suppressed Vegfa expression during pathological conditions. Lack of neuronal and glial SOCS3 resulted in greater phosphorylation and activation of STAT3, which led to increased expression of its gene target Vegfa, and increased endothelial cell proliferation. In summary, SOCS3 in neurons and glial cells inhibited the STAT3-mediated secretion of VEGF from these cells, which suppresses endothelial cell activation, resulting in decreased endothelial cell proliferation and angiogenesis. These results suggest that neuronal and glial cell SOCS3 limits pathological retinal angiogenesis by suppressing VEGF signaling.
Collapse
Affiliation(s)
- Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Meihua Ju
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Zhiqiang Lin
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115, USA
| | - Thomas W Fredrick
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Lucy P Evans
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Katherine T Tian
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Nicholas J Saba
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Peyton C Morss
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - William T Pu
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115, USA. Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Andreas Stahl
- University Eye Hospital Freiburg, Killianstr. 5, Freiburg 79106, Germany
| | - Jean-Sébastien Joyal
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Université de Montréal, Montréal, Québec H3T1C4, Canada
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
40
|
Wilson SS, Wong A, Toupadakis CA, Yellowley CE. Expression of angiopoietin-like protein 4 at the fracture site: Regulation by hypoxia and osteoblastic differentiation. J Orthop Res 2015; 33:1364-73. [PMID: 25864912 DOI: 10.1002/jor.22898] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/10/2015] [Indexed: 02/04/2023]
Abstract
Vascular disruption that occurs as a consequence of bone fracture, leads to hypoxia at the site of damage. Hypoxia regulates the expression of a number of genes that can modulate energy conservation, cell survival, tissue regeneration and angiogenesis. In this study we investigated the expression of Angiopoietin-like 4, an adipocytokine that has additional roles in angiogenesis, at the fracture site. We demonstrate that Angptl4 mRNA expression increased early during fracture healing (day 3) returning close to baseline at day14. In the callus, Angptl4 mRNA was visualized in areas of condensing mesenchymal cells, callus cartilage and was especially high in mineralizing osteoblasts located in areas of new bone formation. In vitro, Angptl4 mRNA expression in osteoblasts increased under hypoxic conditions and in cells treated with the hypoxia mimetic desferrioxamine. Angptl4 levels were strongly induced at day 14 in differentiating MC3T3-E1 osteoblastic cells. Exogenous ANGPTL4 increased expression of Runx2, Spp1, vegfa, and Alp mRNA in differentiating osteoblasts. We suggest that the distribution of Angptl4 in the callus may be driven by hypoxia and that Angptl4 may play a role in osteoblastic differentiation, and possibly angiogenesis via regulation of VEGF. Further studies could reveal a dual role for Angptl4 in angiogenesis and osteogenesis.
Collapse
Affiliation(s)
- Sabrina S Wilson
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, 1285 Veterinary Medicine Drive, Davis, California, 95616
| | - Alice Wong
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, 1285 Veterinary Medicine Drive, Davis, California, 95616
| | - Chrisoula A Toupadakis
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, 1285 Veterinary Medicine Drive, Davis, California, 95616
| | - Clare E Yellowley
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, 1285 Veterinary Medicine Drive, Davis, California, 95616
| |
Collapse
|
41
|
Kwon SH, Shin JP, Kim IT, Park DH. Aqueous Levels of Angiopoietin-like 4 and Semaphorin 3E Correlate with Nonperfusion Area and Macular Volume in Diabetic Retinopathy. Ophthalmology 2015; 122:968-75. [PMID: 25687026 DOI: 10.1016/j.ophtha.2015.01.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 01/07/2015] [Accepted: 01/07/2015] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE To investigate the aqueous levels of angiopoietin-like 4 (ANGPTL4), semaphorin 3E (Sema3E), and vascular endothelial growth factor (VEGF) in patients with diabetic retinopathy and to ascertain their association with diabetic retinopathy phenotypes. DESIGN Prospective, nonrandomized, comparative case series. PARTICIPANTS Of all 104 consecutive patients (104 eyes) who had intravitreal anti-VEGF injections from April 2012 through April 2013 for diabetic macular edema (DME), 51 had severe nonproliferative diabetic retinopathy (NPDR) and 53 had proliferative diabetic retinopathy (PDR). The controls were 54 consecutive nondiabetic patients who had undergone cataract surgery (54 eyes) during the same period. METHODS The ANGPTL4, Sema3E, and VEGF levels in aqueous humor samples obtained before intravitreal injections were measured by enzyme-linked immunosorbent assay. Capillary nonperfusion area (NPA) was calculated from encircled angiography using the 7 standard field images described in the Early Treatment Diabetic Retinopathy Study protocol. Total macular volume (TMV) was measured by spectral-domain optical coherence tomography. MAIN OUTCOME MEASURES Aqueous ANGPTL4, Sema3E, and VEGF levels in severe NPDR, PDR, and control groups and their correlations with each other, NPA, and TMV. RESULTS The severe NPDR and PDR groups had higher aqueous levels of ANGPTL4 and VEGF than the control group (all P < 0.001). The PDR group had higher ANGPTL4 and VEGF levels than the severe NPDR group (both P < 0.001). The aqueous ANGPTL4 levels of all diabetic retinopathy patients correlated positively with NPA (r = 0.820, P = 0.003) and TMV (r = 0.824, P < 0.001). The control group had higher aqueous Sema3E levels than the NPDR and PDR groups (both P < 0.001). Aqueous Sema3E levels correlated negatively with VEGF levels in all subjects (r = -0.57, P = 0.025). CONCLUSIONS The ANGPTL4 may be a candidate target in DME treatment and a biomarker of ischemic-induced retinopathy, including diabetic retinopathy.
Collapse
Affiliation(s)
- Soo Hyun Kwon
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Jae Pil Shin
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - In Taek Kim
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Dong Ho Park
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
42
|
Su EJ, Xin H, Yin P, Dyson M, Coon J, Farrow KN, Mestan KK, Ernst LM. Impaired fetoplacental angiogenesis in growth-restricted fetuses with abnormal umbilical artery doppler velocimetry is mediated by aryl hydrocarbon receptor nuclear translocator (ARNT). J Clin Endocrinol Metab 2015; 100:E30-40. [PMID: 25343232 PMCID: PMC4283004 DOI: 10.1210/jc.2014-2385] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
CONTEXT Fetal growth restriction with abnormal umbilical artery Doppler velocimetry (FGRadv), reflective of elevated fetoplacental vascular resistance, is associated with increased risks of fetal morbidity and mortality even in comparison to those of growth-restricted fetuses with normal placental blood flow. One major cause of this abnormally elevated fetoplacental vascular resistance is the aberrantly formed, thin, elongated villous vessels that are seen in FGRadv placentas. OBJECTIVE The purpose of this study was to determine the role of fetoplacental endothelial cells (ECs) in angiogenesis in normal pregnancies and in those complicated by FGRadv. DESIGN AND PARTICIPANTS Human placental specimens were obtained from FGRadv and gestational age-matched, appropriately grown control pregnancies for EC isolation/culture and for immunohistochemical studies. Additional mechanistic studies were performed on ECs isolated from subjects with term, uncomplicated pregnancies. MAIN OUTCOME MEASURES We evaluated tube formation and differential angiogenic gene expression in FGRadv and control ECs, and we used ECs from uncomplicated pregnancies to further elucidate the molecular mechanisms by which angiogenesis is impaired in FGRadv pregnancies. RESULTS Tube formation assays showed that FGRadv ECs demonstrate fewer branch points and total length compared with those from gestational age-matched controls, and this defect was not rescued by exposure to hypoxia. FGRadv ECs also demonstrated lower aryl hydrocarbon receptor nuclear translocator (ARNT) expression. ARNT knockdown resulted in suppression of key angiogenic genes including vascular endothelial growth factor A expression and led to deficient tube formation. CONCLUSIONS ARNT expression in the placental vasculature mediates key angiogenic expression and fetoplacental EC angiogenesis, and low ARNT expression in FGRadv ECs appears to be a key factor in deficient angiogenesis. This, in turn, results in malformed thin villous vessels that structurally contribute to the abnormally elevated fetoplacental vascular resistance that is associated with high morbidity and mortality in fetal growth restriction.
Collapse
Affiliation(s)
- Emily J Su
- Department of Obstetrics and Gynecology (E.J.S., H.X., P.Y., M.D., J.C.), Division of Maternal-Fetal Medicine and/or Division of Reproductive Science in Medicine, and Department of Pathology (L.M.E.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; and Department of Pediatrics (K.N.F., K.K.M.), Division of Neonatology, Northwestern University Feinberg School of Medicine and Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois 60611
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Ortiz G, Salica JP, Chuluyan EH, Gallo JE. Diabetic retinopathy: could the alpha-1 antitrypsin be a therapeutic option? Biol Res 2014; 47:58. [PMID: 25723058 PMCID: PMC4335423 DOI: 10.1186/0717-6287-47-58] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/13/2014] [Indexed: 02/07/2023] Open
Abstract
Diabetic retinopathy is one of the most important causes of blindness. The underlying mechanisms of this disease include inflammatory changes and remodeling processes of the extracellular-matrix (ECM) leading to pericyte and vascular endothelial cell damage that affects the retinal circulation. In turn, this causes hypoxia leading to release of vascular endothelial growth factor (VEGF) to induce the angiogenesis process. Alpha-1 antitrypsin (AAT) is the most important circulating inhibitor of serine proteases (SERPIN). Its targets include elastase, plasmin, thrombin, trypsin, chymotrypsin, proteinase 3 (PR-3) and plasminogen activator (PAI). AAT modulates the effect of protease-activated receptors (PARs) during inflammatory responses. Plasma levels of AAT can increase 4-fold during acute inflammation then is so-called acute phase protein (APPs). Individuals with low serum levels of AAT could develop disease in lung, liver and pancreas. AAT is involved in extracellular matrix remodeling and inflammation, particularly migration and chemotaxis of neutrophils. It can also suppress nitric oxide (NO) by nitric oxide sintase (NOS) inhibition. AAT binds their targets in an irreversible way resulting in product degradation. The aim of this review is to focus on the points of contact between multiple factors involved in diabetic retinopathy and AAT resembling pleiotropic effects that might be beneficial.
Collapse
Affiliation(s)
- Gustavo Ortiz
- Nanomedicine and Vision Group, Facultad de Ciencias Biomédicas, Universidad Austral, Buenos Aires Pilar, Argentina. .,Ciudad Autónoma de Buenos Aires, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina.
| | - Juan P Salica
- Nanomedicine and Vision Group, Facultad de Ciencias Biomédicas, Universidad Austral, Buenos Aires Pilar, Argentina.
| | - Eduardo H Chuluyan
- Departamento de Farmacología,Ciudad Autónoma de Buenos Aires, Universidad de Buenos Aires, Buenos Aires, Argentina. .,Ciudad Autónoma de Buenos Aires, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina.
| | - Juan E Gallo
- Nanomedicine and Vision Group, Facultad de Ciencias Biomédicas, Universidad Austral, Buenos Aires Pilar, Argentina. .,Ciudad Autónoma de Buenos Aires, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina.
| |
Collapse
|
44
|
Chong HC, Chan JSK, Goh CQ, Gounko NV, Luo B, Wang X, Foo S, Wong MTC, Choong C, Kersten S, Tan NS. Angiopoietin-like 4 stimulates STAT3-mediated iNOS expression and enhances angiogenesis to accelerate wound healing in diabetic mice. Mol Ther 2014; 22:1593-604. [PMID: 24903577 DOI: 10.1038/mt.2014.102] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 05/11/2014] [Indexed: 12/24/2022] Open
Abstract
Impaired wound healing is a major source of morbidity in diabetic patients. Poor outcome has, in part, been related to increased inflammation, poor angiogenesis, and deficiencies in extracellular matrix components. Despite the enormous impact of these chronic wounds, effective therapies are lacking. Here, we showed that the topical application of recombinant matricellular protein angiopoietin-like 4 (ANGPTL4) accelerated wound reepithelialization in diabetic mice, in part, by improving angiogenesis. ANGPTL4 expression is markedly elevated upon normal wound injury. In contrast, ANGPTL4 expression remains low throughout the healing period in diabetic wounds. Exogenous ANGPTL4 modulated several regulatory networks involved in cell migration, angiogenesis, and inflammation, as evidenced by an altered gene expression signature. ANGPTL4 influenced the expression profile of endothelial-specific CD31 in diabetic wounds, returning its profile to that observed in wild-type wounds. We showed ANGPTL4-induced nitric oxide production through an integrin/JAK/STAT3-mediated upregulation of inducible nitric oxide synthase (iNOS) expression in wound epithelia, thus revealing a hitherto unknown mechanism by which ANGPTL4 regulated angiogenesis via keratinocyte-to-endothelial-cell communication. These data show that the replacement of ANGPTL4 may be an effective adjunctive or new therapeutic avenue for treating poor healing wounds. The present finding also confirms that therapeutic angiogenesis remains an attractive treatment modality for diabetic wound healing.
Collapse
Affiliation(s)
- Han Chung Chong
- School of Biological Sciences, Nanyang Technological University, Nanyang Drive, Singapore, Singapore
| | - Jeremy Soon Kiat Chan
- School of Biological Sciences, Nanyang Technological University, Nanyang Drive, Singapore, Singapore
| | - Chi Qin Goh
- School of Biological Sciences, Nanyang Technological University, Nanyang Drive, Singapore, Singapore
| | - Natalia V Gounko
- Institute of Molecular and Cell Biology, Proteos, A*STAR, Singapore, Singapore
| | - Baiwen Luo
- School of Materials Science and Engineering, Nanyang Technological University, Nanyang Avenue, Singapore, Singapore
| | - Xiaoling Wang
- School of Biological Sciences, Nanyang Technological University, Nanyang Drive, Singapore, Singapore
| | - Selin Foo
- School of Biological Sciences, Nanyang Technological University, Nanyang Drive, Singapore, Singapore
| | | | - Cleo Choong
- School of Materials Science and Engineering, Nanyang Technological University, Nanyang Avenue, Singapore, Singapore
| | - Sander Kersten
- Nutrition, Metabolism and Genomics group, Wageningen Univeristy, Wageningen, The Netherlands
| | - Nguan Soon Tan
- 1] School of Biological Sciences, Nanyang Technological University, Nanyang Drive, Singapore, Singapore [2] Institute of Molecular and Cell Biology, Proteos, A*STAR, Singapore, Singapore
| |
Collapse
|
45
|
Inoue T, Kohro T, Tanaka T, Kanki Y, Li G, Poh HM, Mimura I, Kobayashi M, Taguchi A, Maejima T, Suehiro JI, Sugiyama A, Kaneki K, Aruga H, Dong S, Stevens JF, Yamamoto S, Tsutsumi S, Fujita T, Ruan X, Aburatani H, Nangaku M, Ruan Y, Kodama T, Wada Y. Cross-enhancement of ANGPTL4 transcription by HIF1 alpha and PPAR beta/delta is the result of the conformational proximity of two response elements. Genome Biol 2014; 15:R63. [PMID: 24721177 PMCID: PMC4053749 DOI: 10.1186/gb-2014-15-4-r63] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 04/10/2014] [Indexed: 12/11/2022] Open
Abstract
Background Synergistic transcriptional activation by different stimuli has been reported along with a diverse array of mechanisms, but the full scope of these mechanisms has yet to be elucidated. Results We present a detailed investigation of hypoxia-inducible factor (HIF) 1 dependent gene expression in endothelial cells which suggests the importance of crosstalk between the peroxisome proliferator-activated receptor (PPAR) β/δ and HIF signaling axes. A migration assay shows a synergistic interaction between these two stimuli, and we identify angiopoietin-like 4 (ANGPTL4) as a common target gene by using a combination of microarray and ChIP-seq analysis. We profile changes of histone marks at enhancers under hypoxia, PPARβ/δ agonist and dual stimulations and these suggest that the spatial proximity of two response elements is the principal cause of the synergistic transcription induction. A newly developed quantitative chromosome conformation capture assay shows the quantitative change of the frequency of proximity of the two response elements. Conclusions To the best of our knowledge, this is the first report that two different transcription factors cooperate in transcriptional regulation in a synergistic fashion through conformational change of their common target genes.
Collapse
|
46
|
Pirot N, Delpech H, Deleuze V, Dohet C, Courtade-Saïdi M, Basset-Léobon C, Chalhoub E, Mathieu D, Pinet V. Lung endothelial barrier disruption in Lyl1-deficient mice. Am J Physiol Lung Cell Mol Physiol 2014; 306:L775-85. [PMID: 24532287 DOI: 10.1152/ajplung.00200.2013] [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] [Indexed: 12/25/2022] Open
Abstract
Maturation of newly formed vessels is a multistep phenomenon during which functional endothelial barriers are established. Disruption of vessel integrity is an important feature in many physiological and pathological processes. We previously reported that lymphoblastic leukemia-derived sequence 1 (LYL1) is required for the late stages of postnatal angiogenesis to limit the formation of new blood vessels, notably by regulating the activity of the small GTPase Rap1. In this study, we show that LYL1 is also required during the formation of the mature endothelial barrier in the lungs of adult mice. Specifically, LYL1 knockdown in human endothelial cells downregulated the expression of ARHGAP21 and ARHGAP24, which encode two Rho GTPase-activating proteins, and this was correlated with increased RhoA activity and reorganization of the actin cytoskeleton into stress fibers. Importantly, in lungs of Lyl1-deficient mice, both vascular endothelial (VE)-cadherin and p120-catenin were poorly recruited to endothelial adherens junctions, indicative of defective cell-cell junctions. Consistent with this, higher Evans blue dye extravasation, edema, and leukocyte infiltration in the lung parenchyma of Lyl1-/- mice than in wild-type littermates confirmed that lung vascular permeability is constitutively elevated in Lyl1-/- adult mice. Our data show that LYL1 acts as a stabilizing signal for adherens junction formation by operating upstream of VE-cadherin and of the two GTPases Rap1 and RhoA. As increased vascular permeability is a key feature and a major mechanism of acute respiratory distress syndrome, molecules that regulate LYL1 activity could represent additional tools to modify the endothelial barrier permeability.
Collapse
Affiliation(s)
- Nelly Pirot
- Institut de Génétique Moléculaire de Montpellier, UMR 5535, CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Role of Angptl4 in vascular permeability and inflammation. Inflamm Res 2013; 63:13-22. [PMID: 24173241 DOI: 10.1007/s00011-013-0678-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/01/2013] [Accepted: 10/15/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Angptl4 is a secreted protein involved in the regulation of vascular permeability, angiogenesis, and inflammatory responses in different kinds of tissues. Increases of vascular permeability and abnormality changes in angiogenesis contribute to the pathogenesis of tumor metastasis, ischemic-reperfusion injury. Inflammatory response associated with Angptl4 also leads to minimal change glomerulonephritis, wound healing. However, the role of Angptl4 in vascular permeability, angiogenesis, and inflammation is controversy. Hence, an underlying mechanism of Angptl4 in different kind of tissues needs to be further clarified. METHODS Keywords such as angptl4, vascular permeability, angiogenesis, inflammation, and endothelial cells were used in search tool of PUBMED, and then the literatures associated with Angptl4 were founded and read. RESULTS Data have established Angptl4 as the key modulator of both vascular permeability and angiogenesis; furthermore, it may also be related to the progression of metastatic tumors, cardiovascular events, and inflammatory diseases. This view focuses on the recent advances in our understanding of the role of Angptl4 in vascular permeability, angiogenesis, inflammatory signaling and the link between Angptl4 and multiple diseases such as cancer, cardiovascular diseases, diabetic retinopathy, and kidney diseases. CONCLUSIONS Taken together, Angptl4 modulates vascular permeability, angiogenesis, inflammatory signaling, and associated diseases. The use of Angptl4-modulating agents such as certain drugs, food constituents (such as fatty acids), nuclear factor (such as PPARα), and bacteria may treat associated diseases such as tumor metastasis, ischemic-reperfusion injury, inflammation, and chronic low-grade inflammation. However, the diverse physiological functions of Angptl4 in different tissues can lead to potentially deleterious side effects when used as a therapeutic target. In this regard, a better understanding of the underlying mechanisms for Angptl4 in different tissues is necessary.
Collapse
|
48
|
Florea V, Bhagavatula N, Simovic G, Macedo FY, Fock RA, Rodrigues CO. c-Myc is essential to prevent endothelial pro-inflammatory senescent phenotype. PLoS One 2013; 8:e73146. [PMID: 24039874 PMCID: PMC3765198 DOI: 10.1371/journal.pone.0073146] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 07/19/2013] [Indexed: 12/12/2022] Open
Abstract
The proto-oncogene c-Myc is vital for vascular development and promotes tumor angiogenesis, but the mechanisms by which it controls blood vessel growth remain unclear. In the present work we investigated the effects of c-Myc knockdown in endothelial cell functions essential for angiogenesis to define its role in the vasculature. We provide the first evidence that reduction in c-Myc expression in endothelial cells leads to a pro-inflammatory senescent phenotype, features typically observed during vascular aging and pathologies associated with endothelial dysfunction. c-Myc knockdown in human umbilical vein endothelial cells using lentivirus expressing specific anti-c-Myc shRNA reduced proliferation and tube formation. These functional defects were associated with morphological changes, increase in senescence-associated-β-galactosidase activity, upregulation of cell cycle inhibitors and accumulation of c-Myc-deficient cells in G1-phase, indicating that c-Myc knockdown in endothelial cells induces senescence. Gene expression analysis of c-Myc-deficient endothelial cells showed that senescent phenotype was accompanied by significant upregulation of growth factors, adhesion molecules, extracellular-matrix components and remodeling proteins, and a cluster of pro-inflammatory mediators, which include Angptl4, Cxcl12, Mdk, Tgfb2 and Tnfsf15. At the peak of expression of these cytokines, transcription factors known to be involved in growth control (E2f1, Id1 and Myb) were downregulated, while those involved in inflammatory responses (RelB, Stat1, Stat2 and Stat4) were upregulated. Our results demonstrate a novel role for c-Myc in the prevention of vascular pro-inflammatory phenotype, supporting an important physiological function as a central regulator of inflammation and endothelial dysfunction.
Collapse
Affiliation(s)
- Victoria Florea
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Nithya Bhagavatula
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Gordana Simovic
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Francisco Y. Macedo
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Ricardo A. Fock
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Claudia O. Rodrigues
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
49
|
Hypoxic retinal Muller cells promote vascular permeability by HIF-1-dependent up-regulation of angiopoietin-like 4. Proc Natl Acad Sci U S A 2013; 110:E3425-34. [PMID: 23959876 DOI: 10.1073/pnas.1217091110] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Vision loss from ischemic retinopathies commonly results from the accumulation of fluid in the inner retina [macular edema (ME)]. Although the precise events that lead to the development of ME remain under debate, growing evidence supports a role for an ischemia-induced hyperpermeability state regulated, in part, by VEGF. Monthly treatment with anti-VEGF therapies is effective for the treatment of ME but results in a major improvement in vision in a minority of patients, underscoring the need to identify additional therapeutic targets. Using the oxygen-induced retinopathy mouse model for ischemic retinopathy, we provide evidence showing that hypoxic Müller cells promote vascular permeability by stabilizing hypoxia-inducible factor-1α (HIF-1α) and secreting angiogenic cytokines. Blocking HIF-1α translation with digoxin inhibits the promotion of endothelial cell permeability in vitro and retinal edema in vivo. Interestingly, Müller cells require HIF--but not VEGF--to promote vascular permeability, suggesting that other HIF-dependent factors may contribute to the development of ME. Using gene expression analysis, we identify angiopoietin-like 4 (ANGPTL4) as a cytokine up-regulated by HIF-1 in hypoxic Müller cells in vitro and the ischemic inner retina in vivo. ANGPTL4 is critical and sufficient to promote vessel permeability by hypoxic Müller cells. Immunohistochemical analysis of retinal tissue from patients with diabetic eye disease shows that HIF-1α and ANGPTL4 localize to ischemic Müller cells. Our results suggest that ANGPTL4 may play an important role in promoting vessel permeability in ischemic retinopathies and could be an important target for the treatment of ME.
Collapse
|
50
|
Capozzi ME, McCollum GW, Savage SR, Penn JS. Peroxisome proliferator-activated receptor-β/δ regulates angiogenic cell behaviors and oxygen-induced retinopathy. Invest Ophthalmol Vis Sci 2013; 54:4197-207. [PMID: 23716627 DOI: 10.1167/iovs.13-11608] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
PURPOSE To develop new therapies against ocular neovascularization (NV), we tested the effect of peroxisome proliferator-activated receptor-β/δ (PPAR-β/δ) agonism and antagonism on angiogenic behaviors and in human retinal microvascular endothelial cells (HRMEC) and on preretinal NV in rat oxygen-induced retinopathy (OIR). METHODS HRMECs were treated with the PPAR-β/δ agonist GW0742 and the antagonist GSK0660. Messenger RNA levels of a PPAR-β/δ target gene, angiopoietin-like-4 (angptl4) were assayed by qRT-PCR. HRMEC proliferation and tube formation were assayed according to standard protocols. OIR was induced in newborn rats by exposing them to alternating 24-hour episodes of 50% and 10% oxygen for 14 days. OIR rats were treated with GW0742 or GSK0660. Angptl4 protein levels were assessed by ELISA and preretinal NV was quantified by adenosine diphosphatase staining. RESULTS GW0742 significantly increased angptl4 mRNA, and GSK0660 significantly decreased angptl4 mRNA. GW0742 had no effect on HRMEC proliferation, but caused a significant and dose-responsive increase in tube formation. GSK0660 significantly reduced serum-induced HRMEC proliferation and tube formation in a dose-dependent manner. Intravitreal injection of GW0742 significantly increased total retinal Angptl4 protein, but intravitreal injection of GSK0660 had no effect. Intravitreal injection of GW0742 significantly increased retinal NV, as did GW0742 administered by oral gavage. Conversely, both intravitreal injection and intraperitoneal injection of GSK0660 significantly reduced retinal NV. CONCLUSIONS PPAR-β/δ activation exacerbates, and its inhibition reduces, preretinal NV. PPAR-β/δ may regulate preretinal NV through a prodifferentiation/maturation mechanism that depends on Angptl4. Pharmacologic inhibition of PPAR-β/δ may provide a rational basis for therapeutic targeting of ocular NV.
Collapse
Affiliation(s)
- Megan E Capozzi
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | | |
Collapse
|