1
|
Li C, Gou L. FOXA1 exacerbates LPS-induced vascular endothelial cell injury in sepsis by suppressing the transcription of NRP2. Cytotechnology 2024; 76:697-707. [PMID: 39435415 PMCID: PMC11490632 DOI: 10.1007/s10616-024-00647-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/17/2024] [Indexed: 10/23/2024] Open
Abstract
Endothelial dysfunction plays a critical role in the pathogenesis of sepsis. This study aims to explore the effect and mechanism of forkhead box A1 (FOXA1) on vascular endothelial cell injury in sepsis. Human umbilical vein endothelial cells (HUVECs) were stimulated by lipopolysaccharide (LPS). Lactate dehydrogenase (LDH) release, cell viability, apoptosis, and inflammatory factors including IL-1β, TNF-α, and IL-6 were measured using LDH kits, CCK-8 assay, flow cytometry, and ELISA respectively. RT-qPCR or Western blot determined the expression of FOXA1 or neuropilin-2 (NRP2) in cells. The binding between FOXA1 and NRP2 was confirmed using ChIP and dual-luciferase assays. Functional rescue experiments were performed to verify the effect of FOXA1 siRNA or NRP2 siRNA on cell injury. LPS treatment induced endothelial cell injury in a concentration-dependent manner. FOXA1 expression was elevated after LPS treatment. FOXA1 silencing reduced LDH release, enhanced cell viability, suppressed apoptosis, and declined inflammation factors. Mechanistically, FOXA1 bound to the NRP2 promoter to suppress the transcription of NRP2. Functional rescue experiments revealed that knockdown of NRP2 offset the protective effect of knockdown of FOXA1 on cell injury. In conclusion, FOXA1 exacerbates LPS-insulted endothelial cell injury in sepsis by repressing the transcription of NRP2.
Collapse
Affiliation(s)
- Chun Li
- Department of Intensive Care Medicine, Gansu, Second People’s Hospital of Lanzhou City, No. 388, Jingyuan Road, Chengguan District, Lanzhou, 730030 China
| | - Likun Gou
- Department of Intensive Care Medicine, Gansu, Second People’s Hospital of Lanzhou City, No. 388, Jingyuan Road, Chengguan District, Lanzhou, 730030 China
| |
Collapse
|
2
|
Gao RR, Ma LY, Chen JW, Wang YX, Li YY, Zhou ZY, Deng ZH, Zhong J, Shu YH, Liu Y, Chen Q. ATN-161 alleviates caerulein-induced pancreatitis. J Genet Genomics 2024:S1673-8527(24)00262-5. [PMID: 39396744 DOI: 10.1016/j.jgg.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/15/2024]
Abstract
Pancreatitis is a common gastrointestinal disorder that causes hospitalization with significant morbidity and mortality. The mechanistic pathophysiology of pancreatitis is complicated, limiting the discovery of pharmacological intervention methods. Here, we show that the administration of ATN-161, an antagonist of Integrin-α5, significantly mitigates the pathological condition of acute pancreatitis induced by caerulein. We find that CK19-positive pancreatic ductal cells align parallel to blood vessels in the pancreas. In the caerulein-induced acute pancreatitis model, the newly emergent CK19-positive cells are highly vascularized, with a significant increase in vascular density and endothelial cell number. Single-cell RNA sequencing analysis shows that ductal and endothelial cells are intimate interacting partners, suggesting the existence of a ductal-endothelial interface in the pancreas. Pancreatitis dramatically reduces the crosstalk in the ductal-endothelial interface but promotes the Spp-1/Integrin-α5 signaling. Blocking this signaling with ATN-161 significantly reduces acinar-to-ductal metaplasia, pathological angiogenesis, and restores other abnormal defects induced by caerulein. Our work reveals the therapeutic potential of ATN-161 as an uncharacterized pharmacological method to alleviate the symptoms of pancreatitis.
Collapse
Affiliation(s)
- Rong-Rong Gao
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences; NHC Key Laboratory of biotechnology drugs (Shandong Academy of Medical Sciences); Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong 250117, China
| | - Lan-Yue Ma
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China; University of Chinese Academy of Sciences, Beijing 101408, China; China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Jian-Wei Chen
- Institutes of physical science and information technology, Anhui University, Hefei, Anhui 230601, China
| | - Yu-Xiang Wang
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China; University of Chinese Academy of Sciences, Beijing 101408, China; China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Yu-Yan Li
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China; University of Chinese Academy of Sciences, Beijing 101408, China; China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Zi-Yuan Zhou
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518116, China
| | - Zhao-Hua Deng
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China; University of Chinese Academy of Sciences, Beijing 101408, China; China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Jing Zhong
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China; University of Chinese Academy of Sciences, Beijing 101408, China; China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Ya-Hai Shu
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China; China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Yang Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China.
| | - Qi Chen
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences; NHC Key Laboratory of biotechnology drugs (Shandong Academy of Medical Sciences); Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong 250117, China; Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China; China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China; Institutes of physical science and information technology, Anhui University, Hefei, Anhui 230601, China; Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| |
Collapse
|
3
|
Benwell CJ, Johnson RT, Taylor JAGE, Lambert J, Robinson SD. A proteomics approach to isolating neuropilin-dependent α5 integrin trafficking pathways: neuropilin 1 and 2 co-traffic α5 integrin through endosomal p120RasGAP to promote polarised fibronectin fibrillogenesis in endothelial cells. Commun Biol 2024; 7:629. [PMID: 38789481 PMCID: PMC11126613 DOI: 10.1038/s42003-024-06320-4] [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: 12/24/2023] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Integrin trafficking to and from membrane adhesions is a crucial mechanism that dictates many aspects of a cell's behaviour, including motility, polarisation, and invasion. In endothelial cells (ECs), the intracellular traffic of α5 integrin is regulated by both neuropilin 1 (NRP1) and neuropilin 2 (NRP2), yet the redundancies in function between these co-receptors remain unclear. Moreover, the endocytic complexes that participate in NRP-directed traffic remain poorly annotated. Here we identify an important role for the GTPase-activating protein p120RasGAP in ECs, promoting the recycling of α5 integrin from early endosomes. Mechanistically, p120RasGAP enables transit of endocytosed α5 integrin-NRP1-NRP2 complexes to Rab11+ recycling endosomes, promoting cell polarisation and fibronectin (FN) fibrillogenesis. Silencing of both NRP receptors, or p120RasGAP, resulted in the accumulation of α5 integrin in early endosomes, a loss of α5 integrin from surface adhesions, and attenuated EC polarisation. Endothelial-specific deletion of both NRP1 and NRP2 in the postnatal retina recapitulated our in vitro findings, severely impairing FN fibrillogenesis and polarised sprouting. Our data assign an essential role for p120RasGAP during integrin traffic in ECs and support a hypothesis that NRP receptors co-traffic internalised cargoes. Importantly, we utilise comparative proteomics analyses to isolate a comprehensive map of NRP1-dependent and NRP2-dependent α5 integrin interactions in ECs.
Collapse
Affiliation(s)
- Christopher J Benwell
- Food Microbiome and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK.
| | - Robert T Johnson
- Food Microbiome and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - James A G E Taylor
- Food Microbiome and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Jordi Lambert
- Food Microbiome and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
- Section of Cardiorespiratory Medicine, University of Cambridge, VPD Heart & Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge, UK
| | - Stephen D Robinson
- Food Microbiome and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK.
| |
Collapse
|
4
|
Li S, Xu Z, Wang Y, Chen L, Wang X, Zhou Y, Lei D, Zang G, Wang G. Recent advances of mechanosensitive genes in vascular endothelial cells for the formation and treatment of atherosclerosis. Genes Dis 2024; 11:101046. [PMID: 38292174 PMCID: PMC10825297 DOI: 10.1016/j.gendis.2023.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/09/2023] [Accepted: 06/06/2023] [Indexed: 02/01/2024] Open
Abstract
Atherosclerotic cardiovascular disease and its complications are a high-incidence disease worldwide. Numerous studies have shown that blood flow shear has a huge impact on the function of vascular endothelial cells, and it plays an important role in gene regulation of pro-inflammatory, pro-thrombotic, pro-oxidative stress, and cell permeability. Many important endothelial cell mechanosensitive genes have been discovered, including KLK10, CCN gene family, NRP2, YAP, TAZ, HIF-1α, NF-κB, FOS, JUN, TFEB, KLF2/KLF4, NRF2, and ID1. Some of them have been intensively studied, whereas the relevant regulatory mechanism of other genes remains unclear. Focusing on these mechanosensitive genes will provide new strategies for therapeutic intervention in atherosclerotic vascular disease. Thus, this article reviews the mechanosensitive genes affecting vascular endothelial cells, including classical pathways and some newly screened genes, and summarizes the latest research progress on their roles in the pathogenesis of atherosclerosis to reveal effective therapeutic targets of drugs and provide new insights for anti-atherosclerosis.
Collapse
Affiliation(s)
- Shuyu Li
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, National and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Zichen Xu
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, National and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yi Wang
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Lizhao Chen
- Department of Neurosurgery, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing 400042, China
| | - Xiangxiu Wang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, National and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yanghao Zhou
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, National and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Daoxi Lei
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, National and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Guangchao Zang
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Guixue Wang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, National and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| |
Collapse
|
5
|
Szukiewicz D. CX3CL1 (Fractalkine)-CX3CR1 Axis in Inflammation-Induced Angiogenesis and Tumorigenesis. Int J Mol Sci 2024; 25:4679. [PMID: 38731899 PMCID: PMC11083509 DOI: 10.3390/ijms25094679] [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: 03/28/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The chemotactic cytokine fractalkine (FKN, chemokine CX3CL1) has unique properties resulting from the combination of chemoattractants and adhesion molecules. The soluble form (sFKN) has chemotactic properties and strongly attracts T cells and monocytes. The membrane-bound form (mFKN) facilitates diapedesis and is responsible for cell-to-cell adhesion, especially by promoting the strong adhesion of leukocytes (monocytes) to activated endothelial cells with the subsequent formation of an extracellular matrix and angiogenesis. FKN signaling occurs via CX3CR1, which is the only known member of the CX3C chemokine receptor subfamily. Signaling within the FKN-CX3CR1 axis plays an important role in many processes related to inflammation and the immune response, which often occur simultaneously and overlap. FKN is strongly upregulated by hypoxia and/or inflammation-induced inflammatory cytokine release, and it may act locally as a key angiogenic factor in the highly hypoxic tumor microenvironment. The importance of the FKN/CX3CR1 signaling pathway in tumorigenesis and cancer metastasis results from its influence on cell adhesion, apoptosis, and cell migration. This review presents the role of the FKN signaling pathway in the context of angiogenesis in inflammation and cancer. The mechanisms determining the pro- or anti-tumor effects are presented, which are the cause of the seemingly contradictory results that create confusion regarding the therapeutic goals.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
6
|
Lemmens TP, Bröker V, Rijpkema M, Hughes CCW, Schurgers LJ, Cosemans JMEM. Fundamental considerations for designing endothelialized in vitro models of thrombosis. Thromb Res 2024; 236:179-190. [PMID: 38460307 DOI: 10.1016/j.thromres.2024.03.004] [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: 11/18/2023] [Revised: 02/19/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Endothelialized in vitro models for cardiovascular disease have contributed greatly to our current understanding of the complex molecular mechanisms underlying thrombosis. To further elucidate these mechanisms, it is important to consider which fundamental aspects to incorporate into an in vitro model. In this review, we will focus on the design of in vitro endothelialized models of thrombosis. Expanding our understanding of the relation and interplay between the different pathways involved will rely in part on complex models that incorporate endothelial cells, blood, the extracellular matrix, and flow. Importantly, the use of tissue-specific endothelial cells will help in understanding the heterogeneity in thrombotic responses between different vascular beds. The dynamic and complex responses of endothelial cells to different shear rates underlines the importance of incorporating appropriate shear in in vitro models. Alterations in vascular extracellular matrix composition, availability of bioactive molecules, and gradients in concentration and composition of these molecules can all regulate the function of both endothelial cells and perivascular cells. Factors modulating these elements in in vitro models should therefore be considered carefully depending on the research question at hand. As the complexity of in vitro models increases, so can the variability. A bottom-up approach to designing such models will remain an important tool for researchers studying thrombosis. As new techniques are continuously being developed and new pathways are brought to light, research question-dependent considerations will have to be made regarding what aspects of thrombosis to include in in vitro models.
Collapse
Affiliation(s)
- Titus P Lemmens
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Vanessa Bröker
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Minke Rijpkema
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, and Department of Biomedical Engineering, University of California, Irvine, USA
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Judith M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
7
|
Arderiu G, Civit-Urgell A, Díez-Caballero A, Moscatiello F, Ballesta C, Badimon L. Differentiation of Adipose Tissue Mesenchymal Stem Cells into Endothelial Cells Depends on Fat Depot Conditions: Regulation by miRNA. Cells 2024; 13:513. [PMID: 38534357 DOI: 10.3390/cells13060513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024] Open
Abstract
The development of obesity is associated with substantial modulation of adipose tissue (AT) structure. The plasticity of the AT is reflected by its remarkable ability to expand or reduce in size throughout the adult lifespan, which is linked to the development of its vasculature. This increase in AT vasculature could be mediated by the differentiation of adipose tissue-derived stem cells (ASCs) into endothelial cells (ECs) and form new microvasculature. We have already shown that microRNA (miRNA)-145 regulates the differentiation of ASCs into EC-like (ECL) cells. Here, we investigated whether ASCs-differentiation into ECs is governed by a miRNAs signature that depends on fat depot location and /or the metabolic condition produced by obesity. Human ASCs, which were obtained from white AT by surgical procedures from lean and obese patients, were induced to differentiate into ECL cells. We have identified that miRNA-29b-3p in both subcutaneous (s)ASCs and visceral ASCs and miRNA-424-5p and miRNA-378a-3p in subcutaneous (s)ASCs are involved in differentiation into EC-like cells. These miRNAs modulate their pro-angiogenic effects on ASCs by targeting FGFR1, NRP2, MAPK1, and TGF-β2, and the MAPK signaling pathway. We show for the first time that miRNA-29b-3p upregulation contributes to ASCs' differentiation into ECL cells by directly targeting TGFB2 in both sASCs and visceral ASCs. Moreover, our results reveal that, independent of sASCs' origin (obese/lean), the upregulation of miRNA-378a-3p and the downregulation of miRNA-424-5p inhibit MAPK1 and overexpress FGFR1 and NRP2, respectively. In summary, both the adipose depot location and obesity affect the differentiation of resident ASCs through the expression of specific miRNAs.
Collapse
Affiliation(s)
- Gemma Arderiu
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain
- Ciber CV, Instituto Carlos III, 28029 Madrid, Spain
| | - Anna Civit-Urgell
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain
- Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain
| | | | | | - Carlos Ballesta
- Centro Médico Teknon, Grupo Quiron Salut, 08022 Barcelona, Spain
| | - Lina Badimon
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain
- Ciber CV, Instituto Carlos III, 28029 Madrid, Spain
| |
Collapse
|
8
|
Dhupar R, Powers AA, Eisenberg SH, Gemmill RM, Bardawil CE, Udoh HM, Cubitt A, Nangle LA, Soloff AC. Orchestrating Resilience: How Neuropilin-2 and Macrophages Contribute to Cardiothoracic Disease. J Clin Med 2024; 13:1446. [PMID: 38592275 PMCID: PMC10934188 DOI: 10.3390/jcm13051446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 04/10/2024] Open
Abstract
Immunity has evolved to balance the destructive nature of inflammation with wound healing to overcome trauma, infection, environmental insults, and rogue malignant cells. The inflammatory response is marked by overlapping phases of initiation, resolution, and post-resolution remodeling. However, the disruption of these events can lead to prolonged tissue damage and organ dysfunction, resulting long-term disease states. Macrophages are the archetypic phagocytes present within all tissues and are important contributors to these processes. Pleiotropic and highly plastic in their responses, macrophages support tissue homeostasis, repair, and regeneration, all while balancing immunologic self-tolerance with the clearance of noxious stimuli, pathogens, and malignant threats. Neuropilin-2 (Nrp2), a promiscuous co-receptor for growth factors, semaphorins, and integrins, has increasingly been recognized for its unique role in tissue homeostasis and immune regulation. Notably, recent studies have begun to elucidate the role of Nrp2 in both non-hematopoietic cells and macrophages with cardiothoracic disease. Herein, we describe the unique role of Nrp2 in diseases of the heart and lung, with an emphasis on Nrp2 in macrophages, and explore the potential to target Nrp2 as a therapeutic intervention.
Collapse
Affiliation(s)
- Rajeev Dhupar
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Surgical and Research Services, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Amy A. Powers
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Seth H. Eisenberg
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Robert M. Gemmill
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Charles E. Bardawil
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Hannah M. Udoh
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Andrea Cubitt
- aTyr Pharma, San Diego, CA 92121, USA; (A.C.); (L.A.N.)
| | | | - Adam C. Soloff
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Surgical and Research Services, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| |
Collapse
|
9
|
Saha S, Fan F, Alderfer L, Graham F, Hall E, Hanjaya-Putra D. Synthetic hyaluronic acid coating preserves the phenotypes of lymphatic endothelial cells. Biomater Sci 2023; 11:7346-7357. [PMID: 37789798 PMCID: PMC10628678 DOI: 10.1039/d3bm00873h] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/14/2023] [Indexed: 10/05/2023]
Abstract
Lymphatic endothelial cells (LECs) play a critical role in the formation and maintenance of the lymphatic vasculature, which is essential for the immune system, fluid balance, and tissue repair. However, LECs are often difficult to study in vivo and in vitro models that accurately mimic their behaviors and phenotypes are limited. In particular, LECs have been shown to lose their lymphatic markers over time while being cultured in vitro, which reflect their plasticity and heterogeneity in vivo. Since LECs uniquely express lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1), we hypothesized that surface coating with hyaluronic acid (HA) can preserve LEC phenotypes and functionalities. Dopamine conjugated hyaluronic acid (HA-DP) was synthesized with 42% degree of substitution to enable surface modification and conjugation onto standard tissue culture plates. Compared to fibronectin coating and tissue culture plate controls, surface coating with HA-DP was able to preserve lymphatic markers, such as prospero homeobox protein 1 (Prox1), podoplanin (PDPN), and LYVE-1 over several passages in vitro. LECs cultured on HA-DP expressed lower levels of focal adhesion kinase (FAK) and YAP/TAZ, which may be responsible for the maintenance of the lymphatic characteristics. Collectively, the HA-DP coating may provide a novel method for culturing human LECs in vitro toward more representative studies in basic lymphatic biology and lymphatic regeneration.
Collapse
Affiliation(s)
- Sanjoy Saha
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, IN 46556, USA.
| | - Fei Fan
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, IN 46556, USA.
| | - Laura Alderfer
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, IN 46556, USA.
| | - Francine Graham
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, IN 46556, USA
| | - Eva Hall
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, IN 46556, USA.
| | - Donny Hanjaya-Putra
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, IN 46556, USA.
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, IN 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, IN 46556, USA
| |
Collapse
|
10
|
Viúdez-Pareja C, Kreft E, García-Caballero M. Immunomodulatory properties of the lymphatic endothelium in the tumor microenvironment. Front Immunol 2023; 14:1235812. [PMID: 37744339 PMCID: PMC10512957 DOI: 10.3389/fimmu.2023.1235812] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/08/2023] [Indexed: 09/26/2023] Open
Abstract
The tumor microenvironment (TME) is an intricate complex and dynamic structure composed of various cell types, including tumor, stromal and immune cells. Within this complex network, lymphatic endothelial cells (LECs) play a crucial role in regulating immune responses and influencing tumor progression and metastatic dissemination to lymph node and distant organs. Interestingly, LECs possess unique immunomodulatory properties that can either promote or inhibit anti-tumor immune responses. In fact, tumor-associated lymphangiogenesis can facilitate tumor cell dissemination and metastasis supporting immunoevasion, but also, different molecular mechanisms involved in LEC-mediated anti-tumor immunity have been already described. In this context, the crosstalk between cancer cells, LECs and immune cells and how this communication can shape the immune landscape in the TME is gaining increased interest in recent years. In this review, we present a comprehensive and updated report about the immunomodulatory properties of the lymphatic endothelium within the TME, with special focus on primary tumors and tumor-draining lymph nodes. Furthermore, we outline emerging research investigating the potential therapeutic strategies targeting the lymphatic endothelium to enhance anti-tumor immune responses. Understanding the intricate mechanisms involved in LEC-mediated immune modulation in the TME opens up new possibilities for the development of innovative approaches to fight cancer.
Collapse
Affiliation(s)
- Cristina Viúdez-Pareja
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Andalucía Tech, University of Málaga, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| | - Ewa Kreft
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Andalucía Tech, University of Málaga, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| | - Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Andalucía Tech, University of Málaga, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| |
Collapse
|
11
|
Wiggins BG, Wang YF, Burke A, Grunberg N, Vlachaki Walker JM, Dore M, Chahrour C, Pennycook BR, Sanchez-Garrido J, Vernia S, Barr AR, Frankel G, Birdsey GM, Randi AM, Schiering C. Endothelial sensing of AHR ligands regulates intestinal homeostasis. Nature 2023; 621:821-829. [PMID: 37586410 PMCID: PMC10533400 DOI: 10.1038/s41586-023-06508-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/02/2023] [Indexed: 08/18/2023]
Abstract
Endothelial cells line the blood and lymphatic vasculature, and act as an essential physical barrier, control nutrient transport, facilitate tissue immunosurveillance and coordinate angiogenesis and lymphangiogenesis1,2. In the intestine, dietary and microbial cues are particularly important in the regulation of organ homeostasis. However, whether enteric endothelial cells actively sense and integrate such signals is currently unknown. Here we show that the aryl hydrocarbon receptor (AHR) acts as a critical node for endothelial cell sensing of dietary metabolites in adult mice and human primary endothelial cells. We first established a comprehensive single-cell endothelial atlas of the mouse small intestine, uncovering the cellular complexity and functional heterogeneity of blood and lymphatic endothelial cells. Analyses of AHR-mediated responses at single-cell resolution identified tissue-protective transcriptional signatures and regulatory networks promoting cellular quiescence and vascular normalcy at steady state. Endothelial AHR deficiency in adult mice resulted in dysregulated inflammatory responses and the initiation of proliferative pathways. Furthermore, endothelial sensing of dietary AHR ligands was required for optimal protection against enteric infection. In human endothelial cells, AHR signalling promoted quiescence and restrained activation by inflammatory mediators. Together, our data provide a comprehensive dissection of the effect of environmental sensing across the spectrum of enteric endothelia, demonstrating that endothelial AHR signalling integrates dietary cues to maintain tissue homeostasis by promoting endothelial cell quiescence and vascular normalcy.
Collapse
Affiliation(s)
- Benjamin G Wiggins
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
- MRC London Institute of Medical Sciences, London, UK.
| | - Yi-Fang Wang
- MRC London Institute of Medical Sciences, London, UK
| | - Alice Burke
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Nil Grunberg
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Julia M Vlachaki Walker
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Marian Dore
- MRC London Institute of Medical Sciences, London, UK
| | | | - Betheney R Pennycook
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | | | - Santiago Vernia
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Alexis R Barr
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Gad Frankel
- Department of Life Sciences, Imperial College London, London, UK
| | - Graeme M Birdsey
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Anna M Randi
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Chris Schiering
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
- MRC London Institute of Medical Sciences, London, UK.
| |
Collapse
|
12
|
Zhou W, Wang F, Qian X, Luo S, Wang Z, Gao X, Kong X, Zhang J, Chen S. Quercetin protects endothelial function from inflammation induced by localized disturbed flow by inhibiting NRP2 -VEGFC complex. Int Immunopharmacol 2023; 116:109842. [PMID: 36764279 DOI: 10.1016/j.intimp.2023.109842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 01/07/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023]
Abstract
Atherosclerosis is a focal chronic inflammatory disease, the initial pathogenic event of which is endothelial dysfunction, and disturbed flow (DF) is the primary and vital factor underlying endothelial dysfunction. The present research aims to elucidate the mechanism underlying the regulation of Neuropilin (NRP)2 under DF in endothelial cells (ECs) in an inflammatory state. We observed that NRP2 expression was significantly upregulated in DF-stimulated human umbilical vein endothelial cells (HUVECs). Knockdown of NRP2 in HUVECs significantly ameliorated cell inflammation induced by DF. In addition, quercetin inhibited NRP2 expression as well as endothelial inflammation. Animal experiments suggested that NRP2 knockdown or intraperitoneal injection of quercetin affected the expression of inflammation-related genes. Moreover, the upstream transcription factor GATA2 was found to regulate NRP2 transcription by binding to the -1100 to +100 bp region of the NRP2 promoter. Further studies showed that quercetin inhibited NRP2-VEGFC complex formation induced by disturbed flow, although did not inhibit GATA2 expression. These findings suggest that NRP2 plays an important role in promoting inflammation. Quercetin antagonizes atherosclerosis by inhibiting NRP2 and the formation of NRP2-VEGFC complex by inhibiting the inflammatory effects induced by disordered flow.
Collapse
Affiliation(s)
- Wenying Zhou
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University 210029, China
| | - Feng Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University 210029, China
| | - Xuesong Qian
- Department of Cardiology, The First People's Hospital of Zhangjiagang, Zhangjiagang, China
| | - Shuai Luo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University 210029, China
| | - Zhimei Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University 210029, China
| | - Xiaofei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University 210029, China; Department of Cardiology, Nanjing Heart Centre, Nanjing, China
| | - Xiangquan Kong
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University 210029, China; Department of Cardiology, Nanjing Heart Centre, Nanjing, China
| | - Junjie Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University 210029, China; Department of Cardiology, Nanjing Heart Centre, Nanjing, China.
| | - Shaoliang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University 210029, China; Department of Cardiology, Nanjing Heart Centre, Nanjing, China.
| |
Collapse
|
13
|
Steffen BT, Tang W, Lutsey PL, Demmer RT, Selvin E, Matsushita K, Morrison AC, Guan W, Rooney MR, Norby FL, Pankratz N, Couper D, Pankow JS. Proteomic analysis of diabetes genetic risk scores identifies complement C2 and neuropilin-2 as predictors of type 2 diabetes: the Atherosclerosis Risk in Communities (ARIC) Study. Diabetologia 2023; 66:105-115. [PMID: 36194249 PMCID: PMC9742300 DOI: 10.1007/s00125-022-05801-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/15/2022] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Genetic predisposition to type 2 diabetes is well-established, and genetic risk scores (GRS) have been developed that capture heritable liabilities for type 2 diabetes phenotypes. However, the proteins through which these genetic variants influence risk have not been thoroughly investigated. This study aimed to identify proteins and pathways through which type 2 diabetes risk variants may influence pathophysiology. METHODS Using a proteomics data-driven approach in a discovery sample of 7241 White participants in the Atherosclerosis Risk in Communities Study (ARIC) cohort and a replication sample of 1674 Black ARIC participants, we interrogated plasma levels of 4870 proteins and four GRS of specific type 2 diabetes phenotypes related to beta cell function, insulin resistance, lipodystrophy, BMI/blood lipid abnormalities and a composite score of all variants combined. RESULTS Twenty-two plasma proteins were identified in White participants after Bonferroni correction. Of the 22 protein-GRS associations that were statistically significant, 10 were replicated in Black participants and all but one were directionally consistent. In a secondary analysis, 18 of the 22 proteins were found to be associated with prevalent type 2 diabetes and ten proteins were associated with incident type 2 diabetes. Two-sample Mendelian randomisation indicated that complement C2 may be causally related to greater type 2 diabetes risk (inverse variance weighted estimate: OR 1.65 per SD; p=7.0 × 10-3), while neuropilin-2 was inversely associated (OR 0.44 per SD; p=8.0 × 10-3). CONCLUSIONS/INTERPRETATION Identified proteins may represent viable intervention or pharmacological targets to prevent, reverse or slow type 2 diabetes progression, and further research is needed to pursue these targets.
Collapse
Affiliation(s)
- Brian T Steffen
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Pamela L Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Mary R Rooney
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA
| | - Faye L Norby
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Health System, Los Angeles, CA, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, USA
| | - David Couper
- University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
14
|
Integrin receptor trafficking in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:271-302. [PMID: 36813362 DOI: 10.1016/bs.pmbts.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Integrins are a family of 24 different heterodimers that are indispensable for multicellular life. Cell polarity, adhesion and migration are controlled by integrins delivered to the cell surface which in turn is regulated by the exo- and endocytic trafficking of integrins. The deep integration between trafficking and cell signaling determines the spatial and temporal output from any biochemical cue. Integrin trafficking plays a key role in development and many pathological conditions, especially cancer. Several novel regulators of integrin traffic have been discovered in recent times, including a novel class of integrin carrying vesicles, the intracellular nanovesicles (INVs). The tight regulation of trafficking pathways by cell signaling, where kinases phosphorylate key small GTPases in the trafficking pathway enable coordination of cell response to the extracellular milieu. Integrin heterodimer expression and trafficking differ in different tissues and contexts. In this Chapter, we discuss recent studies on integrin trafficking and its contribution to normal physiological and pathophysiological states.
Collapse
|
15
|
Benwell CJ, Johnson RT, Taylor JA, Price CA, Robinson SD. Endothelial VEGFR Coreceptors Neuropilin-1 and Neuropilin-2 Are Essential for Tumor Angiogenesis. CANCER RESEARCH COMMUNICATIONS 2022; 2:1626-1640. [PMID: 36970722 PMCID: PMC10036134 DOI: 10.1158/2767-9764.crc-22-0250] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 10/16/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Neuropilin (NRP) expression is highly correlated with poor outcome in multiple cancer subtypes. As known coreceptors for VEGFRs, core drivers of angiogenesis, past investigations have alluded to their functional roles in facilitating tumorigenesis by promoting invasive vessel growth. Despite this, it remains unclear as to whether NRP1 and NRP2 act in a synergistic manner to enhance pathologic angiogenesis. Here we demonstrate, using NRP1 ECKO , NRP2 ECKO , and NRP1/NRP2 ECKO mouse models, that maximum inhibition of primary tumor development and angiogenesis is achieved when both endothelial NRP1 and NRP2 are targeted simultaneously. Metastasis and secondary site angiogenesis were also significantly inhibited in NRP1/NRP2 ECKO animals. Mechanistic studies revealed that codepleting NRP1 and NRP2 in mouse-microvascular endothelial cells stimulates rapid shuttling of VEGFR-2 to Rab7+ endosomes for proteosomal degradation. Our results highlight the importance of targeting both NRP1 and NRP2 to modulate tumor angiogenesis. Significance The findings presented in this study demonstrate that tumor angiogenesis and growth can be arrested completely by cotargeting endothelial NRP1 and NRP2. We provide new insight into the mechanisms of action regulating NRP-dependent tumor angiogenesis and signpost a novel approach to halt tumor progression.
Collapse
Affiliation(s)
- Christopher J. Benwell
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Robert T. Johnson
- School of Pharmacy, University of East Anglia, Norwich, United Kingdom
| | - James A.G.E. Taylor
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Christopher A. Price
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Stephen D. Robinson
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
- School of Biological Sciences, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
16
|
Gao T, Cao Y, Hu M, Du Y. The activation of TGF-β signaling promotes cell migration and invasion of ectopic endometrium by targeting NRP2. Reprod Biol 2022; 22:100697. [DOI: 10.1016/j.repbio.2022.100697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/06/2022]
|
17
|
Fernández‐Nogales M, López‐Cascales MT, Murcia‐Belmonte V, Escalante A, Fernández‐Albert J, Muñoz‐Viana R, Barco A, Herrera E. Multiomic Analysis of Neurons with Divergent Projection Patterns Identifies Novel Regulators of Axon Pathfinding. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200615. [PMID: 35988153 PMCID: PMC9561852 DOI: 10.1002/advs.202200615] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/30/2022] [Indexed: 06/15/2023]
Abstract
Axon pathfinding is a key step in neural circuits formation. However, the transcriptional mechanisms regulating its progression remain poorly understood. The binary decision of crossing or avoiding the midline taken by some neuronal axons during development represents a robust model to investigate the mechanisms that control the selection of axonal trajectories. Here, to identify novel regulators of axon guidance, this work compares the transcriptome and chromatin occupancy profiles of two neuronal subpopulations, ipsilateral (iRGC) and contralateral retinal ganglion cells (cRGC), with similar functions but divergent axon trajectories. These analyses retrieved a number of genes encoding for proteins not previously implicated in axon pathfinding. In vivo functional experiments confirm the implication of some of these candidates in axonal navigation. Among the candidate genes, γ-synuclein is identified as essential for inducing midline crossing. Footprint and luciferase assays demonstrate that this small-sized protein is regulated by the transcription factor (TF) Pou4f1 in cRGCs. It is also shown that Lhx2/9 are specifically expressed in iRGCs and control a program that partially overlaps with that regulated by Zic2, previously described as essential for iRGC specification. Overall, the analyses identify dozens of new molecules potentially involved in axon guidance and reveal the regulatory logic behind the selection of axonal trajectories.
Collapse
Affiliation(s)
- Marta Fernández‐Nogales
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Maria Teresa López‐Cascales
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Verónica Murcia‐Belmonte
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Augusto Escalante
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Jordi Fernández‐Albert
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Rafael Muñoz‐Viana
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Angel Barco
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Eloísa Herrera
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| |
Collapse
|
18
|
Neuropilin (NRPs) Related Pathological Conditions and Their Modulators. Int J Mol Sci 2022; 23:ijms23158402. [PMID: 35955539 PMCID: PMC9368954 DOI: 10.3390/ijms23158402] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/20/2022] [Accepted: 07/27/2022] [Indexed: 01/08/2023] Open
Abstract
Neuropilin 1 (NRP1) represents one of the two homologous neuropilins (NRP, splice variants of neuropilin 2 are the other) found in all vertebrates. It forms a transmembrane glycoprotein distributed in many human body tissues as a (co)receptor for a variety of different ligands. In addition to its physiological role, it is also associated with various pathological conditions. Recently, NRP1 has been discovered as a coreceptor for the SARS-CoV-2 viral entry, along with ACE2, and has thus become one of the COVID-19 research foci. However, in addition to COVID-19, the current review also summarises its other pathological roles and its involvement in clinical diseases like cancer and neuropathic pain. We also discuss the diversity of native NRP ligands and perform a joint analysis. Last but not least, we review the therapeutic roles of NRP1 and introduce a series of NRP1 modulators, which are typical peptidomimetics or other small molecule antagonists, to provide the medicinal chemistry community with a state-of-the-art overview of neuropilin modulator design and NRP1 druggability assessment.
Collapse
|
19
|
A Circulating Risk Score, Based on Combined Expression of Exo-miR-130a-3p and Fibrinopeptide A, as Predictive Biomarker of Relapse in Resectable Non-Small Cell Lung Cancer Patients. Cancers (Basel) 2022; 14:cancers14143412. [PMID: 35884472 PMCID: PMC9317031 DOI: 10.3390/cancers14143412] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary To date, the five-year survival rate of early stages of non-small cell lung cancer (NSCLC) is still disappointing and reliable prognostic factors are mandatory. Here, we performed in-depth high-throughput analyses of plasma circulating markers, including exosomal microRNAs and peptidome to identify a prognostic score. The miRnome profile selected the Exo-miR-130a-3p as the most overexpressed in relapsed patients. Peptidome analysis identified four progressively more degraded forms of fibrinopeptide A (FpA), which were depleted in relapse patients. Notably, a stepwise algorithm selected Exo-miR-130a-3p and the greatest FpA (2–16) to build a prognostic score, where high-risk patients had 18 months of median disease-free survival. Overexpression of miR-130a-3p cells led to a deregulation of pathways such as angiogenesis as well as the coagulation and metalloprotease, which might be linked to FpA reduction. The risk score integrating circulating markers may help clinicians predict early-stage NSCLC patients who are more likely to relapse after surgery. Abstract To date, the 5-year overall survival rate of 60% for early-stage non-small cell lung cancer (NSCLC) is still unsatisfactory. Therefore, reliable prognostic factors are needed. Growing evidence shows that cancer progression may depend on an interconnection between cancer cells and the surrounding tumor microenvironment; hence, circulating molecules may represent promising markers of cancer recurrence. In order to identify a prognostic score, we performed in-depth high-throughput analyses of plasma circulating markers, including exosomal microRNAs (Exo-miR) and peptides, in 67 radically resected NSCLCs. The miRnome profile selected the Exo-miR-130a-3p as the most overexpressed in relapsed patients. Peptidome analysis identified four progressively more degraded forms of fibrinopeptide A (FpA), which were depleted in progressing patients. Notably, stepwise Cox regression analysis selected Exo-miR-130a-3p and the greatest FpA (2-16) to build a score predictive of recurrence, where high-risk patients had 18 months of median disease-free survival. Moreover, in vitro transfections showed that higher levels of miR-130a-3p lead to a deregulation of pathways involved in metastasis and angiogenesis, including the coagulation process and metalloprotease increase which might be linked to FpA reduction. In conclusion, by integrating circulating markers, the identified risk score may help clinicians predict early-stage NSCLC patients who are more likely to relapse after primary surgery.
Collapse
|
20
|
Islam R, Mishra J, Bodas S, Bhattacharya S, Batra SK, Dutta S, Datta K. Role of Neuropilin-2-mediated signaling axis in cancer progression and therapy resistance. Cancer Metastasis Rev 2022; 41:771-787. [PMID: 35776228 PMCID: PMC9247951 DOI: 10.1007/s10555-022-10048-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 06/16/2022] [Indexed: 12/12/2022]
Abstract
Neuropilins (NRPs) are transmembrane proteins involved in vascular and nervous system development by regulating angiogenesis and axon guidance cues. Several published reports have established their role in tumorigenesis. NRPs are detectable in tumor cells of several cancer types and participate in cancer progression. NRP2 is also expressed in endothelial and immune cells in the tumor microenvironment and promotes functions such as lymphangiogenesis and immune suppression important for cancer progression. In this review, we have taken a comprehensive approach to discussing various aspects of NRP2-signaling in cancer, including its regulation, functional significance in cancer progression, and how we could utilize our current knowledge to advance the studies and target NRP2 to develop effective cancer therapies.
Collapse
Affiliation(s)
- Ridwan Islam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Juhi Mishra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanika Bodas
- Department of Molecular Genetics and Cell Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Sreyashi Bhattacharya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Samikshan Dutta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
21
|
Luo S, Wang F, Chen S, Chen A, Wang Z, Gao X, Kong X, Zuo G, Zhou W, Gu Y, Ge Z, Zhang J. NRP2 promotes atherosclerosis by upregulating PARP1 expression and enhancing low shear stress-induced endothelial cell apoptosis. FASEB J 2022; 36:e22079. [PMID: 35028975 DOI: 10.1096/fj.202101250rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 01/13/2023]
Abstract
Atherosclerosis-related cardiovascular diseases are leading causes of mortality worldwide, characterized by the development of endothelial cell dysfunction, increased oxidized low-density lipoprotein uptake by macrophages, and the ensuing formation of atherosclerotic plaque. Local blood flow patterns cause uneven atherosclerotic lesion distribution, and endothelial dysfunction caused by disturbed flow is an early step in the development of atherosclerosis. The present research aims to elucidate the mechanism underlying the regulation of Neuropilin 2 (NRP2) under low shear stress (LSS) in the atheroprone phenotype of endothelial cells. We observed that NRP2 expression was significantly upregulated in LSS-stimulated human umbilical vein endothelial cells (HUVECs) and in mouse aortic endothelial cells. Knockdown of NRP2 in HUVECs significantly ameliorated cell apoptosis induced by LSS. Conversely, overexpression of NRP2 had the opposite effect on HUVEC apoptosis. Animal experiments suggest that NRP2 knockdown markedly mitigated the development of atherosclerosis in Apoe-/- mice. Mechanistically, NRP2 knockdown and overexpression regulated PARP1 protein expression in the condition of LSS, which in turn affected the expression of apoptosis-related genes. Moreover, the upstream transcription factor GATA2 was found to regulate NRP2 expression in the progression of atherosclerosis. These findings suggest that NRP2 plays an essential proatherosclerotic role through the regulation of cell apoptosis, and the results reveal that NRP2 is a promising therapeutic target for the treatment of atherosclerotic disorders.
Collapse
Affiliation(s)
- Shuai Luo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Feng Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Siyu Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Aiqun Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zhimei Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaofei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Cardiology, Nanjing Heart Centre, Nanjing, China
| | - Xiangquan Kong
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Guangfeng Zuo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Wenying Zhou
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Gu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zhen Ge
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Junjie Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Cardiology, Nanjing Heart Centre, Nanjing, China
| |
Collapse
|
22
|
Taylor JAGE, Benwell CJ, Robinson SD. Using Immortalized Endothelial Cells to Study the Roles of Adhesion Molecules in VEGF-Induced Signaling. Methods Mol Biol 2022; 2475:133-141. [PMID: 35451754 DOI: 10.1007/978-1-0716-2217-9_9] [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] [Indexed: 10/18/2022]
Abstract
The ability to study the role of specific genes in endothelial cell biology is made possible by our ability to modulate their expression through siRNA or knockout technologies. However, many in vitro protocols, particularly those of a biochemical nature, require large numbers of endothelial cells. These types of analyses are encumbered by the need to repeatedly produce and characterize primary endothelial cell cultures and can be greatly facilitated by the use of immortalized microvascular endothelial cells. However, we have found that the manipulation of gene expression in these cells is not always straight forward. Here we describe how we alter gene expression in polyoma middle T antigen immortalized microvascular endothelial cells isolated from wild-type and genetically modified mice to study the role of cell adhesion molecules in downstream assays.
Collapse
Affiliation(s)
- James A G E Taylor
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Christopher J Benwell
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Stephen D Robinson
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK.
| |
Collapse
|
23
|
A novel circUBR4/miR-491-5p/NRP2 ceRNA network regulates oxidized low-density lipoprotein-induced proliferation and migration in vascular smooth muscle cells. J Cardiovasc Pharmacol 2021; 79:512-522. [PMID: 34935701 DOI: 10.1097/fjc.0000000000001204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 11/28/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Vascular smooth muscle cells (VSMCs) play critical roles in the progression of atherosclerosis. Circular RNA (circRNA) ubiquitin protein ligase E3 component n-recognin 4 (circUBR4) has been shown to regulate VSMC migration and proliferation. Here, we sought to identify the mechanism in the regulation of circUBR4. CircUBR4, microRNA (miR)-491-5p and Neuropilin-2 (NRP2) were quantified by quantitative real-time PCR (qRT-PCR) and western blot. Cell proliferation was evaluated by Cell Counting Kit-8 (CCK-8) and 5-Ethynyl-2'-Deoxyuridine (EDU) assays. Cell migration was examined by wound-healing and transwell invasion assays. The direct relationship between miR-491-5p and circUBR4 or NRP2 was validated by dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. Our data indicated that in VSMCs, ox-LDL induced circUBR4 expression. Silencing endogenous circUBR4 attenuated VSMC proliferation and migration induced by ox-LDL. Mechanistically, circUBR4 targeted miR-491-5p by pairing to miR-491-5p. Moreover, miR-491-5p was identified as a downstream mediator of circUBR4 function in ox-LDL-treated VSMCs. NRP2 was a direct target of miR-491-5p, and circUBR4 acted as a competing endogenous RNA (ceRNA) for miR-491-5p to regulate NRP2 expression. Additionally, NRP2 was a functionally downstream effector of miR-491-5p in regulating ox-LDL-evoked VSMC proliferation and migration. Our findings identify a new ceRNA network, the circUBR4/miR-491-5p/NRP2 axis, for the regulation of circUBR4 in VSMC migration and proliferation.
Collapse
|
24
|
Mascarenhas JB, Gaber AA, Larrinaga TM, Mayfield R, Novak S, Camp SM, Gregorio C, Jacobson JR, Cress AE, Dudek SM, Garcia JGN. EVL is a novel focal adhesion protein involved in the regulation of cytoskeletal dynamics and vascular permeability. Pulm Circ 2021; 11:20458940211049002. [PMID: 34631011 PMCID: PMC8493322 DOI: 10.1177/20458940211049002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022] Open
Abstract
Increases in lung vascular permeability is a cardinal feature of inflammatory disease and represents an imbalance in vascular contractile forces and barrier-restorative forces, with both forces highly dependent upon the actin cytoskeleton. The current study investigates the role of Ena-VASP-like (EVL), a member of the Ena-VASP family known to regulate the actin cytoskeleton, in regulating vascular permeability responses and lung endothelial cell barrier integrity. Utilizing changes in transendothelial electricial resistance (TEER) to measure endothelial cell barrier responses, we demonstrate that EVL expression regulates endothelial cell responses to both sphingosine-1-phospate (S1P), a vascular barrier-enhancing agonist, and to thrombin, a barrier-disrupting stimulus. Total internal reflection fluorescence demonstrates that EVL is present in endothelial cell focal adhesions and impacts focal adhesion size, distribution, and the number of focal adhesions generated in response to S1P and thrombin challenge, with the focal adhesion kinase (FAK) a key contributor in S1P-stimulated EVL-transduced endothelial cell but a limited role in thrombin-induced focal adhesion rearrangements. In summary, these data indicate that EVL is a focal adhesion protein intimately involved in regulation of cytoskeletal responses to endothelial cell barrier-altering stimuli. Keywords: cytoskeleton, vascular barrier, sphingosine-1-phosphate, thrombin, focal adhesion kinase (FAK), Ena-VASP like protein (EVL), cytoskeletal regulatory protein
Collapse
Affiliation(s)
| | - Amir A Gaber
- Department of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Tania M Larrinaga
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Rachel Mayfield
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Stefanie Novak
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Sara M Camp
- Department of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Carol Gregorio
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Jeffrey R Jacobson
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Anne E Cress
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Steven M Dudek
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Joe G N Garcia
- Department of Medicine, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
25
|
Colotti G, Failla CM, Lacal PM, Ungarelli M, Ruffini F, Di Micco P, Orecchia A, Morea V. Neuropilin-1 is required for endothelial cell adhesion to soluble vascular endothelial growth factor receptor 1. FEBS J 2021; 289:183-198. [PMID: 34252269 PMCID: PMC9290910 DOI: 10.1111/febs.16119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 05/27/2021] [Accepted: 07/12/2021] [Indexed: 12/22/2022]
Abstract
Neuropilin‐1 (NRP‐1) is a semaphorin receptor involved in neuron guidance, and a co‐receptor for selected isoforms of the vascular endothelial growth factor (VEGF) family. NRP‐1 binding to several VEGF‐A isoforms promotes growth factor interaction with VEGF receptor (VEGFR)‐2, increasing receptor phosphorylation. Additionally, NRP‐1 directly interacts with VEGFR‐1, but this interaction competes with NRP‐1 binding to VEGF‐A165 and does not enhance VEGFR‐1 activation. In this work, we investigated in detail the role of NRP‐1 interaction with the soluble isoform of VEGFR‐1 (sVEGFR‐1) in angiogenesis. sVEGFR‐1 acts both as a decoy receptor for VEGFs and as an extracellular matrix protein directly binding to α5β1 integrin on endothelial cells. By combining cell adhesion assays and surface plasmon resonance experiments on purified proteins, we found that sVEGFR‐1/NRP‐1 interaction is required both for α5β1 integrin binding to sVEGFR‐1 and for endothelial cell adhesion to a sVEGFR‐1‐containing matrix. We also found that a previously reported anti‐angiogenic peptide (Flt2‐11), which maps in the second VEGFR‐1 Ig‐like domain, specifically binds NRP‐1 and inhibits NRP‐1/sVEGFR‐1 interaction, a process that likely contributes to its anti‐angiogenic activity. In view of potential translational applications, we developed a five‐residue‐long peptide, derived from Flt2‐11, which has the same ability as the parent Flt2‐11 peptide to inhibit cell adhesion to, and migration towards, sVEGFR‐1. Therefore, the Flt2‐5 peptide represents a potential anti‐angiogenic compound per se, as well as an attractive lead for the development of novel angiogenesis inhibitors acting with a different mechanism with respect to currently used therapeutics, which interfere with VEGF‐A165 binding.
Collapse
Affiliation(s)
- Gianni Colotti
- Institute of Molecular Biology and Pathology (IBPM) of the National Research Council (CNR), Rome, Italy
| | | | | | | | | | - Patrizio Di Micco
- Department of Biochemical Sciences 'A. Rossi Fanelli', Sapienza' University of Rome, Italy
| | - Angela Orecchia
- Laboratory of Molecular and Cell Biology, IDI-IRCCS, Rome, Italy
| | - Veronica Morea
- Institute of Molecular Biology and Pathology (IBPM) of the National Research Council (CNR), Rome, Italy
| |
Collapse
|
26
|
Barrantes FJ. The unfolding palette of COVID-19 multisystemic syndrome and its neurological manifestations. Brain Behav Immun Health 2021; 14:100251. [PMID: 33842898 PMCID: PMC8019247 DOI: 10.1016/j.bbih.2021.100251] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 02/07/2023] Open
Abstract
Although our current knowledge of the pathophysiology of COVID-19 is still fragmentary, the information so far accrued on the tropism and life cycle of its etiological agent SARS-CoV-2, together with the emerging clinical data, suffice to indicate that the severe acute pulmonary syndrome is the main, but not the only manifestation of COVID-19. Necropsy studies are increasingly revealing underlying endothelial vasculopathies in the form of micro-haemorrhages and micro-thrombi. Intertwined with defective antiviral responses, dysregulated coagulation mechanisms, abnormal hyper-inflammatory reactions and responses, COVID-19 is disclosing a wide pathophysiological palette. An additional property in categorising the disease is the combination of tissue (e.g. neuro- and vasculo-tropism) with organ tropism, whereby the virus preferentially attacks certain organs with highly developed capillary beds, such as the lungs, gastrointestinal tract, kidney and brain. These multiple clinical presentations confirm that the acute respiratory syndrome as described initially is increasingly unfolding as a more complex nosological entity, a multiorgan syndrome of systemic breadth. The neurological manifestations of COVID-19, the focus of this review, reflect this manifold nature of the disease.
Collapse
Affiliation(s)
- Francisco J. Barrantes
- Institute of Biomedical Research (BIOMED), UCA-CONICET, Av. Alicia Moreau de Justo 1600, C1107AFF, Buenos Aires, Argentina
| |
Collapse
|
27
|
Harman JL, Sayers J, Chapman C, Pellet-Many C. Emerging Roles for Neuropilin-2 in Cardiovascular Disease. Int J Mol Sci 2020; 21:E5154. [PMID: 32708258 PMCID: PMC7404143 DOI: 10.3390/ijms21145154] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular disease, the leading cause of death worldwide, is predominantly associated with atherosclerosis. Atherosclerosis is a chronic inflammatory disease characterised by the narrowing of large to medium-sized arteries due to a build-up of plaque. Atherosclerotic plaque is comprised of lipids, extracellular matrix, and several cell types, including endothelial, immune, and vascular smooth muscle cells. Such narrowing of the blood vessels can itself restrict blood flow to vital organs but most severe clinical complications, including heart attacks and strokes, occur when lesions rupture, triggering the blood to clot and obstructing blood flow further down the vascular tree. To circumvent such obstructions, percutaneous coronary intervention or bypass grafts are often required; however, re-occlusion of the treated artery frequently occurs. Neuropilins (NRPs), a multifunctional family of cell surface co-receptors, are expressed by endothelial, immune, and vascular smooth muscle cells and are regulators of numerous signalling pathways within the vasculature. Here, we review recent studies implicating NRP2 in the development of occlusive vascular diseases and discuss how NRP2 could be targeted for therapeutic intervention.
Collapse
Affiliation(s)
- Jennifer L Harman
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Jacob Sayers
- University College London, Division of Medicine, Rayne Building, University Street, London WC1E 6JF, UK
| | - Chey Chapman
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Caroline Pellet-Many
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| |
Collapse
|
28
|
Dumond A, Pagès G. Neuropilins, as Relevant Oncology Target: Their Role in the Tumoral Microenvironment. Front Cell Dev Biol 2020; 8:662. [PMID: 32766254 PMCID: PMC7380111 DOI: 10.3389/fcell.2020.00662] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/01/2020] [Indexed: 12/18/2022] Open
Abstract
Angiogenesis is one of the key mechanisms involved in tumor growth and metastatic dissemination. The vascular endothelial growth factor (VEGF) and its receptors (VEGFR) represent one of the major signaling pathways which mediates angiogenesis. The VEGF/VEGFR axis was intensively targeted by monoclonal antibodies or by tyrosine kinase inhibitors to destroy the tumor vascular network. By inhibiting oxygen and nutrient supply, this strategy was supposed to cure cancers. However, despite a lengthening of the progression free survival in several types of tumors including colon, lung, breast, kidney, and ovarian cancers, modest improvements in overall survival were reported. Anti-angiogenic therapies targeting VEGF/VEGFR are still used in colon and ovarian cancer and remain reference treatments for renal cell carcinoma. Although the concept of inhibiting angiogenesis remains relevant, new targets need to be discovered to improve the therapeutic index of anti-VEGF/VEGFR. Neuropilin 1 and 2 (NRP1/2), initially described as neuronal receptors, stimulate angiogenesis, lymphangiogenesis and immune tolerance. Moreover, overexpression of NRPs in several tumors is synonymous of patients' shorter survival. This article aims to overview the different roles of NRPs in cells constituting the tumor microenvironment to highlight the therapeutic relevance of their targeting.
Collapse
Affiliation(s)
- Aurore Dumond
- Medical Biology Department, Centre Scientifique de Monaco, Monaco, Monaco
| | - Gilles Pagès
- Medical Biology Department, Centre Scientifique de Monaco, Monaco, Monaco.,Inserm U1081, CNRS UMR 7284, Centre Antoine Lacassagne, Institut de Recherche sur le Cancer et le Vieillissement de Nice, Université Côte d'Azur, Nice, France
| |
Collapse
|