1
|
Wang Y, Liu G, Wang J, Zhou P, Zhang L, Liu Q, Zhou J. NRP1 downregulation correlates with enhanced ILC2 responses during IL-33 challenge. Immunology 2024; 172:226-234. [PMID: 38409805 DOI: 10.1111/imm.13769] [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: 07/10/2023] [Accepted: 02/08/2024] [Indexed: 02/28/2024] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) play critical roles in driving the pathogenesis of allergic airway inflammation. The mechanisms underlying the regulation of ILC2s remain to be fully understood. Here, we identified neuropilin-1 (NRP1) as a surface marker of ILC2s in response to IL-33 stimulation. NRP1 was abundantly expressed in ILC2s from lung under steady state, which was significantly reduced upon IL-33 stimulation. ILC2s with high expression of NRP1 (NRP1high) displayed lower response to IL-33, as compared with NRP1low ILC2s. Transcriptional profiling and flow cytometric analysis showed that downregulation of AKT-mTOR signalling participated in the diminished functionality of NRP1high ILC2s. These observations revealed a potential role of NRP1 in ILC2s responses under allergic inflammatory condition.
Collapse
Affiliation(s)
- Ying Wang
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Gaoyu Liu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jianye Wang
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Pan Zhou
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lijuan Zhang
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qiang Liu
- Tianjin Medical University General Hospital, Tianjin Neurological Institute, Tianjin Institute of Immunology, Tianjin, China
| | - Jie Zhou
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
2
|
Ceci C, Lacal PM, Barbaccia ML, Mercuri NB, Graziani G, Ledonne A. The VEGFs/VEGFRs system in Alzheimer's and Parkinson's diseases: Pathophysiological roles and therapeutic implications. Pharmacol Res 2024; 201:107101. [PMID: 38336311 DOI: 10.1016/j.phrs.2024.107101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
The vascular endothelial growth factors (VEGFs) and their cognate receptors (VEGFRs), besides their well-known involvement in physiological angiogenesis/lymphangiogenesis and in diseases associated to pathological vessel formation, play multifaceted functions in the central nervous system (CNS). In addition to shaping brain development, by controlling cerebral vasculogenesis and regulating neurogenesis as well as astrocyte differentiation, the VEGFs/VEGFRs axis exerts essential functions in the adult brain both in physiological and pathological contexts. In this article, after describing the physiological VEGFs/VEGFRs functions in the CNS, we focus on the VEGFs/VEGFRs involvement in neurodegenerative diseases by reviewing the current literature on the rather complex VEGFs/VEGFRs contribution to the pathogenic mechanisms of Alzheimer's (AD) and Parkinson's (PD) diseases. Thereafter, based on the outcome of VEGFs/VEGFRs targeting in animal models of AD and PD, we discuss the factual relevance of pharmacological VEGFs/VEGFRs modulation as a novel and potential disease-modifying approach for these neurodegenerative pathologies. Specific VEGFRs targeting, aimed at selective VEGFR-1 inhibition, while preserving VEGFR-2 signal transduction, appears as a promising strategy to hit the molecular mechanisms underlying AD pathology. Moreover, therapeutic VEGFs-based approaches can be proposed for PD treatment, with the aim of fine-tuning their brain levels to amplify neurotrophic/neuroprotective effects while limiting an excessive impact on vascular permeability.
Collapse
Affiliation(s)
- Claudia Ceci
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Maria Luisa Barbaccia
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Neurology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; IRCCS Santa Lucia Foundation, Department of Experimental Neuroscience, Rome, Italy; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Grazia Graziani
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Ada Ledonne
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; IRCCS Santa Lucia Foundation, Department of Experimental Neuroscience, Rome, Italy; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
3
|
Chikh A, Raimondi C. Endothelial Neuropilin-1: a multifaced signal transducer with an emerging role in inflammation and atherosclerosis beyond angiogenesis. Biochem Soc Trans 2024; 52:137-150. [PMID: 38323651 PMCID: PMC10903451 DOI: 10.1042/bst20230329] [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: 10/24/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 02/08/2024]
Abstract
Neuropilin-1 (NRP1) is a transmembrane glycoprotein expressed by several cell types including, neurons, endothelial cells (ECs), smooth muscle cells, cardiomyocytes and immune cells comprising macrophages, dendritic cells and T cell subsets. Since NRP1 discovery in 1987 as an adhesion molecule in the frog nervous system, more than 2300 publications on PubMed investigated the function of NRP1 in physiological and pathological contexts. NRP1 has been characterised as a coreceptor for class 3 semaphorins and several members of the vascular endothelial growth factor (VEGF) family. Because the VEGF family is the main regulator of blood and lymphatic vessel growth in addition to promoting neurogenesis, neuronal patterning, neuroprotection and glial growth, the role of NRP1 in these biological processes has been extensively investigated. It is now established that NRP1 promotes the physiological growth of new vessels from pre-existing ones in the process of angiogenesis. Furthermore, several studies have shown that NRP1 mediates signalling pathways regulating pathological vascular growth in ocular neovascular diseases and tumour development. Less defined are the roles of NRP1 in maintaining the function of the quiescent established vasculature in an adult organism. This review will focus on the opposite roles of NRP1 in regulating transforming growth factor β signalling pathways in different cell types, and on the emerging role of endothelial NRP1 as an atheroprotective, anti-inflammatory factor involved in the response of ECs to shear stress.
Collapse
Affiliation(s)
- Anissa Chikh
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, U.K
| | - Claudio Raimondi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, U.K
| |
Collapse
|
4
|
van der Veen I, Heredero Berzal A, Koster C, ten Asbroek ALMA, Bergen AA, Boon CJF. The Road towards Gene Therapy for X-Linked Juvenile Retinoschisis: A Systematic Review of Preclinical Gene Therapy in Cell-Based and Rodent Models of XLRS. Int J Mol Sci 2024; 25:1267. [PMID: 38279267 PMCID: PMC10816913 DOI: 10.3390/ijms25021267] [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: 12/22/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
X-linked juvenile retinoschisis (XLRS) is an early-onset progressive inherited retinopathy affecting males. It is characterized by abnormalities in the macula, with formation of cystoid retinal cavities, frequently accompanied by splitting of the retinal layers, impaired synaptic transmission of visual signals, and associated loss of visual acuity. XLRS is caused by loss-of-function mutations in the retinoschisin gene located on the X chromosome (RS1, MIM 30083). While proof-of-concept studies for gene augmentation therapy have been promising in in vitro and rodent models, clinical trials in XLRS patients have not been successful thus far. We performed a systematic literature investigation using search strings related to XLRS and gene therapy in in vivo and in vitro models. Three rounds of screening (title/abstract, full text and qualitative) were performed by two independent reviewers until consensus was reached. Characteristics related to study design and intervention were extracted from all studies. Results were divided into studies using (1) viral and (2) non-viral therapies. All in vivo rodent studies that used viral vectors were assessed for quality and risk of bias using the SYRCLE's risk-of-bias tool. Studies using alternative and non-viral delivery techniques, either in vivo or in vitro, were extracted and reviewed qualitatively, given the diverse and dispersed nature of the information. For in-depth analysis of in vivo studies using viral vectors, outcome data for optical coherence tomography (OCT), immunohistopathology and electroretinography (ERG) were extracted. Meta-analyses were performed on the effect of recombinant adeno-associated viral vector (AAV)-mediated gene augmentation therapies on a- and b-wave amplitude as well as the ratio between b- and a-wave amplitudes (b/a-ratio) extracted from ERG data. Subgroup analyses and meta-regression were performed for model, dose, age at injection, follow-up time point and delivery method. Between-study heterogeneity was assessed with a Chi-square test of homogeneity (I2). We identified 25 studies that target RS1 and met our search string. A total of 19 of these studies reported rodent viral methods in vivo. Six of the 25 studies used non-viral or alternative delivery methods, either in vitro or in vivo. Of these, five studies described non-viral methods and one study described an alternative delivery method. The 19 aforementioned in vivo studies were assessed for risk of bias and quality assessments and showed inconsistency in reporting. This resulted in an unclear risk of bias in most included studies. All 19 studies used AAVs to deliver intact human or murine RS1 in rodent models for XLRS. Meta-analyses of a-wave amplitude, b-wave amplitude, and b/a-ratio showed that, overall, AAV-mediated gene augmentation therapy significantly ameliorated the disease phenotype on these parameters. Subgroup analyses and meta-regression showed significant correlations between b-wave amplitude effect size and dose, although between-study heterogeneity was high. This systematic review reiterates the high potential for gene therapy in XLRS, while highlighting the importance of careful preclinical study design and reporting. The establishment of a systematic approach in these studies is essential to effectively translate this knowledge into novel and improved treatment alternatives.
Collapse
Affiliation(s)
- Isa van der Veen
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Andrea Heredero Berzal
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Céline Koster
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Anneloor L. M. A. ten Asbroek
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Arthur A. Bergen
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Camiel J. F. Boon
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Ophthalmology, Leiden University Medical Center, Leiden University, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
5
|
Saleki K, Alijanizadeh P, Azadmehr A. Is neuropilin-1 the neuroimmune initiator of multi-system hyperinflammation in COVID-19? Biomed Pharmacother 2023; 167:115558. [PMID: 37748412 DOI: 10.1016/j.biopha.2023.115558] [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: 07/09/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023] Open
Abstract
A major immunopathological feature of Coronavirus disease-2019 (COVID-19) is excessive inflammation in the form of "cytokine storm". The storm is characterized by injurious levels of cytokines which form a complicated network damaging different organs, including the lungs and the brain. The main starter of "cytokine network" hyperactivation in COVID-19 has not been discovered yet. Neuropilins (NRPs) are transmembrane proteins that act as neuronal guidance and angiogenesis modulators. The crucial function of NRPs in forming the nervous and vascular systems has been well-studied. NRP1 and NRP2 are the two identified homologs of NRP. NRP1 has been shown as a viral entry pathway for SARS-CoV2, which facilitates neuroinvasion by the virus within the central or peripheral nervous systems. These molecules directly interact with various COVID-19-related molecules, such as specific regions of the spike protein (major immune element of SARS-CoV2), vascular endothelial growth factor (VEGF) receptors, VEGFR1/2, and ANGPTL4 (regulator of vessel permeability and integrity). NRPs mainly play a role in hyperinflammatory injury of the CNS and lungs, and also the liver, kidney, pancreas, and heart in COVID-19 patients. New findings have suggested NRPs good candidates for pharmacotherapy of COVID-19. However, therapeutic targeting of NRP1 in COVID-19 is still in the preclinical phase. This review presents the implications of NRP1 in multi-organ inflammation-induced injury by SARS-CoV2 and provides insights for NRP1-targeting treatments for COVID-19 patients.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences(SBMU), Tehran, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Abbas Azadmehr
- Immunology Department, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
6
|
Bosseboeuf E, Chikh A, Chaker AB, Mitchell TP, Vignaraja D, Rajendrakumar R, Khambata RS, Nightingale TD, Mason JC, Randi AM, Ahluwalia A, Raimondi C. Neuropilin-1 interacts with VE-cadherin and TGFBR2 to stabilize adherens junctions and prevent activation of endothelium under flow. Sci Signal 2023; 16:eabo4863. [PMID: 37220183 PMCID: PMC7614756 DOI: 10.1126/scisignal.abo4863] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/03/2023] [Indexed: 05/25/2023]
Abstract
Linear and disturbed flow differentially regulate gene expression, with disturbed flow priming endothelial cells (ECs) for a proinflammatory, atheroprone expression profile and phenotype. Here, we investigated the role of the transmembrane protein neuropilin-1 (NRP1) in ECs exposed to flow using cultured ECs, mice with an endothelium-specific knockout of NRP1, and a mouse model of atherosclerosis. We demonstrated that NRP1 was a constituent of adherens junctions that interacted with VE-cadherin and promoted its association with p120 catenin, stabilizing adherens junctions and inducing cytoskeletal remodeling in alignment with the direction of flow. We also showed that NRP1 interacted with transforming growth factor-β (TGF-β) receptor II (TGFBR2) and reduced the plasma membrane localization of TGFBR2 and TGF-β signaling. NRP1 knockdown increased the abundance of proinflammatory cytokines and adhesion molecules, resulting in increased leukocyte rolling and atherosclerotic plaque size. These findings describe a role for NRP1 in promoting endothelial function and reveal a mechanism by which NRP1 reduction in ECs may contribute to vascular disease by modulating adherens junction signaling and promoting TGF-β signaling and inflammation.
Collapse
Affiliation(s)
- Emy Bosseboeuf
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Anissa Chikh
- Molecular and Clinical Sciences Research Institute, St. George’s, University of London, London SW17 0RE, UK
| | - Ahmed Bey Chaker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Tom P. Mitchell
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre for Microvascular Research, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Dhilakshani Vignaraja
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Ridhi Rajendrakumar
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Rayomand S. Khambata
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Thomas D. Nightingale
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre for Microvascular Research, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Justin C. Mason
- Vascular Sciences, National Heart & Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Anna M. Randi
- Vascular Sciences, National Heart & Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Amrita Ahluwalia
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Claudio Raimondi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
7
|
Abstract
Semaphorin 3A is a secreted glycoprotein, which was originally identified as axon guidance factor in the neuronal system, but it also possesses immunoregulatory properties. Here, the effect of semaphorin 3A on T-lymphocytes, myeloid dendritic cells and macrophages is systematically analyzed on the bases of all publications available in the literature for 20 years. Expression of semaphorin 3A receptors – neuropilin-1 and plexins A – in these cells is described in details. The data obtained on human and murine cells is described comparatively. A comprehensive overview of the interaction of semaphorin 3A with mononuclear phagocyte system is presented for the first time. Semaphorin 3A signaling mostly results in changes of the cytoskeletal machinery and cellular morphology that regulate pathways involved in migration, adhesion, and cell–cell cooperation of immune cells. Accumulating evidence indicates that this factor is crucially involved in various phases of immune responses, including initiation phase, antigen presentation, effector T cell function, inflammation phase, macrophage activation, and polarization. In recent years, interest in this field has increased significantly because semaphorin 3A is associated with many human diseases and therefore can be used as a target for their treatment. Its involvement in the immune responses is important to study, because semaphorin 3A and its receptors turn to be a promising new therapeutic tools to be applied in many autoimmune, allergic, and oncology diseases.
Collapse
Affiliation(s)
- Ekaterina P Kiseleva
- Federal State Budgetary Scientific Institution "Institute of Experimental Medicine", St. Petersburg, 197376, Russia.
- Mechnikov North-Western State Medical University, St. Petersburg, 195067, Russia
| | - Kristina V Rutto
- Federal State Budgetary Scientific Institution "Institute of Experimental Medicine", St. Petersburg, 197376, Russia.
| |
Collapse
|
8
|
Wu S, Huang Y, Huang X, Dai X. Lipopolysaccharide Accelerates Neuropilin-1 Protein Degradation by Activating the Large GTPase Dynamin-1 in Macrophages. Inflammation 2022; 45:1162-1173. [DOI: 10.1007/s10753-021-01610-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/23/2022]
|
9
|
Regulation of granulosa cell functions through NRP-1 mediated internalization of follicular fluid non-exosomal miR-210. Cell Tissue Res 2021; 386:649-660. [PMID: 34599690 DOI: 10.1007/s00441-021-03524-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/07/2021] [Indexed: 10/20/2022]
Abstract
Crosstalk between follicular fluid (FF) and granulosa cells (GCs) plays a vital role in the regulation of folliculogenesis, ensuring regular reproductive cycle in mammals. This crosstalk is primarily mediated by hormones and signaling molecules, such as cytokines and chemokines. Recently, extracellular microRNAs (miRNAs) have gained a lot of attention in cell-to-cell communication. Extracellular miRNA transportation occurs through exosomes, a kind of micro-vesicles produced from almost all cells. However, the mode of non-exosomal miRNA internalization is not much studied. In the present study, we explored the role of neuropilin-1 (NRP-1) as a receptor in internalizing FF non-exosomal miRNAs in GCs. We first confirmed the expression of NRP-1 in GCs during follicular development followed by its role in the internalization of miR-210, a non-exosomal miRNA. This study showed that incubation of GCs with a non-exosomal fraction of FF increased the content of miR-210 in GCs as compared to their control. To illustrate the role of NRP-1 as a receptor, NRP-1 was knockdown using siRNA. Silencing experimental results showed a significant decrease in uptake of miR-210 in NRP-1 knockdown GCs. Furthermore, downstream expression analysis of miR-210 target genes (CYP19A1, PCNA, and EFNA3) also confirmed the NRP-1 mediated miR-210 internalization. Results of the present study clearly demonstrated that FF non-exosomal miR-210 can be internalized through the NRP-1 receptor. Furthermore, differential expression of NRP-1 in GCs suggests its role in follicular development. Overall, these findings suggest that FF non-exosomal miRNA plays an important role in GC functions and female reproduction.
Collapse
|
10
|
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
|
11
|
Chekol Abebe E, Mengie Ayele T, Tilahun Muche Z, Asmamaw Dejenie T. Neuropilin 1: A Novel Entry Factor for SARS-CoV-2 Infection and a Potential Therapeutic Target. Biologics 2021; 15:143-152. [PMID: 33986591 PMCID: PMC8110213 DOI: 10.2147/btt.s307352] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022]
Abstract
The novel coronavirus disease 2019 (COVID-19) pandemic is severely challenging the healthcare systems and economies of the world, which urgently demand vaccine and therapy development to combat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Hence, advancing our understanding of the comprehensive entry mechanisms of SARS-CoV-2, especially the host factors that facilitate viral infection, is crucial for the discovery of effective vaccines and antiviral drugs. SARS-CoV-2 has previously been documented to reach cells by binding with ACE2 and CD147 receptors in host cells that interact with the spike (S) protein of SARS-CoV-2. A novel entry factor, called neuropilin 1(NRP1), has recently been discovered as a co-receptor facilitating the entry of SARS-CoV-2. NRP1 is a single-pass transmembrane glycoprotein widely distributed throughout the tissues of the body and acts as a multifunctional co-receptor to bind with different ligand proteins and play diverse physiological roles as well as pathological and therapeutic roles in different clinical conditions/diseases, including COVID-19. The current review, therefore, briefly provides the overview of SARS-CoV-2 entry mechanisms, the structure of NRP1, and their roles in health and various diseases, as well as extensively discusses the current understanding of the potential implication of NRP1 in SARS-CoV-2 entry and COVID-19 treatment.
Collapse
Affiliation(s)
- Endeshaw Chekol Abebe
- Department of Medical Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Teklie Mengie Ayele
- Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Zelalem Tilahun Muche
- Department of Medical Physiology, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Tadesse Asmamaw Dejenie
- Department of Medical Biochemistry, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
12
|
Silva TP, Sousa-Luís R, Fernandes TG, Bekman EP, Rodrigues CAV, Vaz SH, Moreira LM, Hashimura Y, Jung S, Lee B, Carmo-Fonseca M, Cabral JMS. Transcriptome profiling of human pluripotent stem cell-derived cerebellar organoids reveals faster commitment under dynamic conditions. Biotechnol Bioeng 2021; 118:2781-2803. [PMID: 33871054 DOI: 10.1002/bit.27797] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/30/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022]
Abstract
Human-induced pluripotent stem cells (iPSCs) have great potential for disease modeling. However, generating iPSC-derived models to study brain diseases remains a challenge. In particular, the ability to recapitulate cerebellar development in vitro is still limited. We presented a reproducible and scalable production of cerebellar organoids by using the novel single-use Vertical-Wheel bioreactors, in which functional cerebellar neurons were obtained. Here, we evaluate the global gene expression profiles by RNA sequencing (RNA-seq) across cerebellar differentiation, demonstrating a faster cerebellar commitment in this novel dynamic differentiation protocol. Furthermore, transcriptomic profiles suggest a significant enrichment of extracellular matrix (ECM) in dynamic-derived cerebellar organoids, which can better mimic the neural microenvironment and support a consistent neuronal network. Thus, an efficient generation of organoids with cerebellar identity was achieved for the first time in a continuous process using a dynamic system without the need of organoids encapsulation in ECM-based hydrogels, allowing the possibility of large-scale production and application in high-throughput processes. The presence of factors that favors angiogenesis onset was also detected in dynamic conditions, which can enhance functional maturation of cerebellar organoids. We anticipate that large-scale production of cerebellar organoids may help developing models for drug screening, toxicological tests, and studying pathological pathways involved in cerebellar degeneration.
Collapse
Affiliation(s)
- Teresa P Silva
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rui Sousa-Luís
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Evguenia P Bekman
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Carlos A V Rodrigues
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Sandra H Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal, Portugal
| | - Leonilde M Moreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | | | | | - Brian Lee
- PBS Biotech, Camarillo, California, USA
| | - Maria Carmo-Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
13
|
Neuropilin 1 Regulation of Vascular Permeability Signaling. Biomolecules 2021; 11:biom11050666. [PMID: 33947161 PMCID: PMC8146136 DOI: 10.3390/biom11050666] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/24/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022] Open
Abstract
The vascular endothelium acts as a selective barrier to regulate macromolecule exchange between the blood and tissues. However, the integrity of the endothelium barrier is compromised in an array of pathological settings, including ischemic disease and cancer, which are the leading causes of death worldwide. The resulting vascular hyperpermeability to plasma molecules as well as leukocytes then leads to tissue damaging edema formation and inflammation. The vascular endothelial growth factor A (VEGFA) is a potent permeability factor, and therefore a desirable target for impeding vascular hyperpermeability. However, VEGFA also promotes angiogenesis, the growth of new blood vessels, which is required for reperfusion of ischemic tissues. Moreover, edema increases interstitial pressure in poorly perfused tumors, thereby affecting the delivery of therapeutics, which could be counteracted by stimulating the growth of new functional blood vessels. Thus, targets must be identified to accurately modulate the barrier function of blood vessels without affecting angiogenesis, as well as to develop more effective pro- or anti-angiogenic therapies. Recent studies have shown that the VEGFA co-receptor neuropilin 1 (NRP1) could be playing a fundamental role in steering VEGFA-induced responses of vascular endothelial cells towards angiogenesis or vascular permeability. Moreover, NRP1 is involved in mediating permeability signals induced by ligands other than VEGFA. This review therefore focuses on current knowledge on the role of NRP1 in the regulation of vascular permeability signaling in the endothelium to provide an up-to-date landscape of the current knowledge in this field.
Collapse
|
14
|
Nrp-1 Mediated Plasmatic Ago2 Binding miR-21a-3p Internalization: A Novel Mechanism for miR-21a-3p Accumulation in Renal Tubular Epithelial Cells during Sepsis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2370253. [PMID: 32923478 PMCID: PMC7453242 DOI: 10.1155/2020/2370253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 12/13/2022]
Abstract
The mechanism underlying sepsis-associated acute kidney injury (SAKI), which is an independent risk factor for sepsis-associated death, is unclear. A previous study indicates that during sepsis miR-21a-3p accumulates in renal tubular epithelial cells (TECs) as the mediator of inflammation and mediates TEC malfunction by manipulating its metabolism. However, the specific mechanism responsible for the accumulation of miR-21a-3p in TECs during sepsis is unrevealed. In this study, a cecal ligation and puncture- (CLP-) induced sepsis rat model and rat TEC line were used to elucidate the mechanism. Firstly, miR-21a-3p and Ago2 levels were found out to increase in both plasma and TECs during sepsis, and the increase of intracellular Ago2 and miR-21a-3p could be mitigated when Ago2 was either inactivated or downregulated in septic plasma. Moreover, membrane Nrp-1 expression of TECs was increased significantly during sepsis and Nrp-1 knockdown also mitigated the rise of both the intracellular Ago2 and miR-21a-3p levels in TECs incubated with septic plasma. Furthermore, it was revealed that Ago2 can be internalized by TECs mediated with Nrp-1 and this process had no effect on the intracellular content of miR-21a-3p. Both Ago2 and miR-21a-3p could bind to TECs derived Nrp-1 directly. Finally, it was determined that miR-21a-3p was internalized by TECs via Nrp-1 and Ago2 facilitated this process. Taken together, it can be concluded from our results that Ago2 binding miR-21a-3p from septic plasma can be actively internalized by TECs via Nrp-1 mediated cell internalization, and this mechanism is crucial for the rise of intracellular miR-21a-3p content of TECs during sepsis. These findings will improve our understanding of the mechanisms underlying SAKI and aid in developing novel therapeutic strategies.
Collapse
|
15
|
Sema3a-Nrp1 Signaling Mediates Fast-Twitch Myofiber Specificity of Tw2 + Cells. Dev Cell 2019; 51:89-98.e4. [PMID: 31474563 DOI: 10.1016/j.devcel.2019.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/03/2019] [Accepted: 08/02/2019] [Indexed: 12/11/2022]
Abstract
We previously identified a unique population of interstitial muscle progenitors, marked by expression of the Twist2 transcription factor, which fuses specifically to type IIb/x fast-twitch myofibers. Tw2+ progenitors are distinct from satellite cells, a muscle progenitor that expresses Pax7 and contributes to all myofiber types. Through RNA sequencing and immunofluorescence, we identify the membrane receptor, Nrp1, as a marker of Tw2+ cells but not Pax7+ cells. We also found that Sema3a, a chemorepellent ligand for Nrp1, is expressed by type I and IIa myofibers but not IIb myofibers. Using stripe migration assays, chimeric cell-cell fusion assays, and a Sema3a transgenic mouse model, we identify Sema3a-Nrp1 signaling as a major mechanism for Tw2+ cell fiber-type specificity. Our findings reveal an extracellular signaling mechanism whereby a cell-surface receptor for a chemorepellent confers specificity of intercellular fusion of a specific muscle progenitor with its target tissue.
Collapse
|
16
|
Battin C, De Sousa Linhares A, Paster W, Isenman DE, Wahrmann M, Leitner J, Zlabinger GJ, Steinberger P, Hofer J. Neuropilin-1 Acts as a Receptor for Complement Split Products. Front Immunol 2019; 10:2209. [PMID: 31572401 PMCID: PMC6753332 DOI: 10.3389/fimmu.2019.02209] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/02/2019] [Indexed: 01/07/2023] Open
Abstract
Complement split products (CSPs), such as the fragments C4d and C3d, which are generated as a consequence of complement regulatory processes, are established markers for disease activity in autoimmunity or antibody-mediated graft rejection. Since immunoglobulin-like transcript 4 (ILT4) was previously shown to interact with soluble CSPs, but not with CSPs covalently-bound to target surfaces following classical complement activation, the present study aimed to identify novel cellular receptors interacting with covalently-deposited CSPs. By applying an unbiased screening approach using a cDNA mammalian expression library generated from human monocyte-derived dendritic cells and probed with recombinant human C4d, we identified neuropilin-1 (NRP1) as a novel receptor for C4d, C3d, and iC3b. NRP1, a highly conserved type 1 transmembrane protein, plays important roles in the development of the nervous and cardiovascular system as well as in tumorigenesis through interaction with its established binding partners, such as vascular endothelial growth factor (VEGF) and semaphorin 3A (Sema3A). NRP1 is also expressed on immune cells and serves as a marker for murine Tregs. Although NRP1 contains domains homologous to ones found in some complement proteins, it has not been linked to the complement system. We demonstrate that binding of C4d to NRP1 expressing cells was dose-dependent and saturable, and had a KD value of 0.71 μM. Importantly, and in contrast to ILT4, NRP1 interacted with CSPs that were covalently bound to target surfaces in the course of complement activation, therefore representing a classical complement receptor. The binding site of CSPs was mapped to the b1 domain of the coagulation factor V/VIII homology domain of NRP1. Taken together, our results demonstrate a novel role for NRP1 as a receptor for CSPs deposited on surfaces during complement activation. Further work is required to elucidate the functional consequences of the NRP1-CSP interactions in immunity.
Collapse
Affiliation(s)
- Claire Battin
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Annika De Sousa Linhares
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Paster
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.,Department of Clinical Cell Biology and FACS Core Unit, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - David E Isenman
- Departments of Biochemistry and Immunology, University of Toronto, Toronto, ON, Canada
| | - Markus Wahrmann
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University Vienna, Vienna, Austria
| | - Judith Leitner
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerhard J Zlabinger
- Division of Clinical and Experimental Immunology, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Johannes Hofer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University Vienna, Vienna, Austria
| |
Collapse
|
17
|
Erskine L, François U, Denti L, Joyce A, Tillo M, Bruce F, Vargesson N, Ruhrberg C. VEGF-A and neuropilin 1 (NRP1) shape axon projections in the developing CNS via dual roles in neurons and blood vessels. Development 2017; 144:2504-2516. [PMID: 28676569 PMCID: PMC5536872 DOI: 10.1242/dev.151621] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/30/2017] [Indexed: 12/11/2022]
Abstract
Visual information is relayed from the eye to the brain via retinal ganglion cell (RGC) axons. Mice lacking NRP1 or NRP1-binding VEGF-A isoforms have defective RGC axon organisation alongside brain vascular defects. It is not known whether axonal defects are caused exclusively by defective VEGF-A signalling in RGCs or are exacerbated by abnormal vascular morphology. Targeted NRP1 ablation in RGCs with a Brn3bCre knock-in allele reduced axonal midline crossing at the optic chiasm and optic tract fasciculation. In contrast, Tie2-Cre-mediated endothelial NRP1 ablation induced axon exclusion zones in the optic tracts without impairing axon crossing. Similar defects were observed in Vegfa120/120 and Vegfa188/188 mice, which have vascular defects as a result of their expression of single VEGF-A isoforms. Ectopic midline vascularisation in endothelial Nrp1 and Vegfa188/188 mutants caused additional axonal exclusion zones within the chiasm. As in vitro and in vivo assays demonstrated that vessels do not repel axons, abnormally large or ectopically positioned vessels are likely to present physical obstacles to axon growth. We conclude that proper axonal wiring during brain development depends on the precise molecular control of neurovascular co-patterning. Summary: NRP1 plays a dual role in retinal ganglion cells and in vascular endothelial cells to organise axons along the optic pathway between the mouse retina and diencephalon.
Collapse
Affiliation(s)
- Lynda Erskine
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Urielle François
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Andy Joyce
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Miguel Tillo
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Freyja Bruce
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Neil Vargesson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| |
Collapse
|
18
|
Wu L, Leng D, Cun D, Foged C, Yang M. Advances in combination therapy of lung cancer: Rationales, delivery technologies and dosage regimens. J Control Release 2017; 260:78-91. [PMID: 28527735 DOI: 10.1016/j.jconrel.2017.05.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 05/13/2017] [Accepted: 05/16/2017] [Indexed: 01/30/2023]
Abstract
Lung cancer is a complex disease caused by a multitude of genetic and environmental factors. The progression of lung cancer involves dynamic changes in the genome and a complex network of interactions between cancer cells with multiple, distinct cell types that form tumors. Combination therapy using different pharmaceuticals has been proven highly effective due to the ability to affect multiple cellular pathways involved in the disease progression. However, the currently used drug combination designs are primarily based on empirical clinical studies, and little attention has been given to dosage regimens, i.e. how administration routes, onsets, and durations of the combinations influence the therapeutic outcome. This is partly because combination therapy is challenged by distinct physicochemical properties and in vivo pharmacokinetics/pharmacodynamics of the individual pharmaceuticals, including small molecule drugs and biopharmaceuticals, which make the optimization of dosing and administration schedule challenging. This article reviews the recent advances in the design and development of combinations of pharmaceuticals for the treatment of lung cancer. Focus is primarily on rationales for the selection of specific combination therapies for lung cancer treatment, and state of the art of delivery technologies and dosage regimens for the combinations, tested in preclinical and clinical trials.
Collapse
Affiliation(s)
- Lan Wu
- Wuya College of Innovation, Shenyang Pharmaceutical University, 110016 Shenyang, China
| | - Donglei Leng
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Dongmei Cun
- Wuya College of Innovation, Shenyang Pharmaceutical University, 110016 Shenyang, China
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Mingshi Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, 110016 Shenyang, China; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
19
|
Abstract
Hisato Kondoh and Harukazu Nakamura look back at the life and career of their mentor Tokindo S. Okada, a pioneer of Japanese developmental biology.
Collapse
Affiliation(s)
- Hisato Kondoh
- Kyoto Sangyo University Faculty of Life Sciences, Kamigamo Motoyama, Kita-ku, Kyoto 603-8555, Japan
| | - Harukazu Nakamura
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| |
Collapse
|
20
|
Sano N, Shimogawa T, Sakaguchi H, Ioroi Y, Miyawaki Y, Morizane A, Miyamoto S, Takahashi J. Enhanced Axonal Extension of Subcortical Projection Neurons Isolated from Murine Embryonic Cortex using Neuropilin-1. Front Cell Neurosci 2017; 11:123. [PMID: 28507510 PMCID: PMC5410565 DOI: 10.3389/fncel.2017.00123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/12/2017] [Indexed: 12/12/2022] Open
Abstract
The cerebral cortical tissue of murine embryo and pluripotent stem cell (PSC)-derived neurons can survive in the brain and extend axons to the spinal cord. For efficient cell integration to the corticospinal tract (CST) after transplantation, the induction or selection of cortical motor neurons is important. However, precise information about the appropriate cell population remains unclear. To address this issue, we isolated cells expressing Neuropilin-1 (NRP1), a major axon guidance molecule receptor during the early developmental stage, from E14.5 mouse embryonic frontal cortex by fluorescence-activated cell sorting. Aggregates of NRP1+ cells gradually expressed subcortical projection neuron markers, Ctip2 and VGluT1, and axon guidance molecule receptors, Robo1 and deleted in colorectal calcinoma (Dcc), in vitro, suggesting that they contained early-stage subcortical projection neurons. We transplanted NRP1+ cells into the frontal cortex of P2 neonatal mice. Compared with grafts derived from NRP1− or unsorted cells, those derived from NRP1+ cells extended a larger number of axons to the spinal cord along the CST. Our data suggest that sorting NRP1+ cells from the embryonic cerebral cortex enriches subcortical projection neurons to reconstruct the CST.
Collapse
Affiliation(s)
- Noritaka Sano
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto UniversityKyoto, Japan.,Department of Neurosurgery, Kyoto University School of MedicineKyoto, Japan
| | - Takafumi Shimogawa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto UniversityKyoto, Japan.,Department of Neurosurgery, Graduate School of Medical sciences, Kyushu UniversityFukuoka, Japan
| | - Hideya Sakaguchi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto UniversityKyoto, Japan
| | - Yoshihiko Ioroi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto UniversityKyoto, Japan.,Department of Neurosurgery, National Hospital Organization Himeji Medical CenterHyogo, Japan
| | - Yoshifumi Miyawaki
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto UniversityKyoto, Japan
| | - Asuka Morizane
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto UniversityKyoto, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University School of MedicineKyoto, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto UniversityKyoto, Japan.,Department of Neurosurgery, Kyoto University School of MedicineKyoto, Japan
| |
Collapse
|
21
|
Kukreja S, Gautam P, Saxena R, Saxena M, Udaykumar N, Kumar A, Bhatt R, Kumar V, Sen J. Identification of novel candidate regulators of retinotectal map formation through transcriptional profiling of the chick optic tectum. J Comp Neurol 2017; 525:459-477. [PMID: 27410778 DOI: 10.1002/cne.24073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 07/06/2016] [Accepted: 07/08/2016] [Indexed: 11/06/2022]
Abstract
Information from the retina is carried along the visual pathway with accuracy and spatial conservation as a result of topographically mapped axonal connections. The optic tectum in the midbrain is the primary region to which retinal ganglion cells project their axons in the chick. The two primary axes of the retina project independently onto the tectum using different sets of guidance cues to give rise to the retinotectal map. Specificity of the map is determined by attractive or repulsive interactions between molecular tags that are distributed in gradients in the retina and the tectum. Despite several studies, knowledge of the retinotectal guidance molecules is far from being complete. We screened for all molecules that are expressed differentially along the anterior-posterior and medial-lateral axes of the chick tectum using microarray based transcriptional profiling and identified several novel candidate retinotectal guidance molecules. Two such genes, encoding Wnt5a and Raldh2, the synthesizing enzymes for retinoic acid, were further analyzed for their function as putative regulators of retinotectal map formation. Wnt5a and retinoic acid were found to exhibit differential effects on the growth of axons from retinal explants derived from different quadrants of the retina. This screen also yielded a large number of genes expressed in a lamina-specific manner in the tectum, which may have other roles in tectal development. J. Comp. Neurol. 525:459-477, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shweta Kukreja
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, 208016, India.,Shweta Kukreja is now at the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605
| | - Pratibha Gautam
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, 208016, India.,Pratibha Gautam is now at the Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Richa Saxena
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, 208016, India.,Richa Saxena is now at the Central Drug Research Institute, Jankipuram, Lucknow, Uttar Pradesh, 226031, India
| | - Monika Saxena
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, 208016, India
| | - Niveda Udaykumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, 208016, India
| | - Aditi Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, 208016, India
| | - Ritesh Bhatt
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, 208016, India
| | - Vidur Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, 208016, India
| | - Jonaki Sen
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, 208016, India
| |
Collapse
|
22
|
Telley L, Cadilhac C, Cioni JM, Saywell V, Jahannault-Talignani C, Huettl RE, Sarrailh-Faivre C, Dayer A, Huber AB, Ango F. Dual Function of NRP1 in Axon Guidance and Subcellular Target Recognition in Cerebellum. Neuron 2016; 91:1276-1291. [PMID: 27618676 DOI: 10.1016/j.neuron.2016.08.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 02/05/2016] [Accepted: 07/30/2016] [Indexed: 11/17/2022]
Abstract
Subcellular target recognition in the CNS is the culmination of a multiple-step program including axon guidance, target recognition, and synaptogenesis. In cerebellum, basket cells (BCs) innervate the soma and axon initial segment (AIS) of Purkinje cells (PCs) to form the pinceau synapse, but the underlying mechanisms remain incompletely understood. Here, we demonstrate that neuropilin-1 (NRP1), a Semaphorin receptor expressed in BCs, controls both axonal guidance and subcellular target recognition. We show that loss of Semaphorin 3A function or specific deletion of NRP1 in BCs alters the stereotyped organization of BC axon and impairs pinceau synapse formation. Further, we identified NRP1 as a trans-synaptic binding partner of the cell adhesion molecule neurofascin-186 (NF186) expressed in the PC AIS during pinceau synapse formation. These findings identify a dual function of NRP1 in both axon guidance and subcellular target recognition in the construction of GABAergic circuitry.
Collapse
Affiliation(s)
- Ludovic Telley
- Department of Neurobiology, Institut de Génomique Fonctionnelle, CNRS, UMR5203, 34090 Montpellier, France; INSERM, U1191, 34094 Montpellier, France; Université de Montpellier, 34090 Montpellier, France; Department of Basic Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | - Christelle Cadilhac
- Department of Neurobiology, Institut de Génomique Fonctionnelle, CNRS, UMR5203, 34090 Montpellier, France; INSERM, U1191, 34094 Montpellier, France; Université de Montpellier, 34090 Montpellier, France; Department of Basic Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | - Jean-Michel Cioni
- Department of Neurobiology, Institut de Génomique Fonctionnelle, CNRS, UMR5203, 34090 Montpellier, France; INSERM, U1191, 34094 Montpellier, France; Université de Montpellier, 34090 Montpellier, France; Department of Physiology Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| | - Veronique Saywell
- Department of Neurobiology, Institut de Génomique Fonctionnelle, CNRS, UMR5203, 34090 Montpellier, France; INSERM, U1191, 34094 Montpellier, France; Université de Montpellier, 34090 Montpellier, France
| | - Céline Jahannault-Talignani
- Department of Neurobiology, Institut de Génomique Fonctionnelle, CNRS, UMR5203, 34090 Montpellier, France; INSERM, U1191, 34094 Montpellier, France; Université de Montpellier, 34090 Montpellier, France
| | - Rosa E Huettl
- Institute of Developmental Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | | | - Alexandre Dayer
- Department of Basic Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland; Department of Mental Health and Psychiatry, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | - Andrea B Huber
- Institute of Developmental Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Fabrice Ango
- Department of Neurobiology, Institut de Génomique Fonctionnelle, CNRS, UMR5203, 34090 Montpellier, France; INSERM, U1191, 34094 Montpellier, France; Université de Montpellier, 34090 Montpellier, France.
| |
Collapse
|
23
|
Wang Y, Cao Y, Mangalam AK, Guo Y, LaFrance-Corey RG, Gamez JD, Atanga PA, Clarkson BD, Zhang Y, Wang E, Angom RS, Dutta K, Ji B, Pirko I, Lucchinetti CF, Howe CL, Mukhopadhyay D. Neuropilin-1 modulates interferon-γ-stimulated signaling in brain microvascular endothelial cells. J Cell Sci 2016; 129:3911-3921. [PMID: 27591257 DOI: 10.1242/jcs.190702] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/31/2016] [Indexed: 02/06/2023] Open
Abstract
Inflammatory response of blood-brain barrier (BBB) endothelial cells plays an important role in pathogenesis of many central nervous system inflammatory diseases, including multiple sclerosis; however, the molecular mechanism mediating BBB endothelial cell inflammatory response remains unclear. In this study, we first observed that knockdown of neuropilin-1 (NRP1), a co-receptor of several structurally diverse ligands, suppressed interferon-γ (IFNγ)-induced C-X-C motif chemokine 10 expression and activation of STAT1 in brain microvascular endothelial cells in a Rac1-dependent manner. Moreover, endothelial-specific NRP1-knockout mice, VECadherin-Cre-ERT2/NRP1flox/flox mice, showed attenuated disease progression during experimental autoimmune encephalomyelitis, a mouse neuroinflammatory disease model. Detailed analysis utilizing histological staining, quantitative PCR, flow cytometry and magnetic resonance imaging demonstrated that deletion of endothelial NRP1 suppressed neuron demyelination, altered lymphocyte infiltration, preserved BBB function and decreased activation of the STAT1-CXCL10 pathway. Furthermore, increased expression of NRP1 was observed in endothelial cells of acute multiple sclerosis lesions. Our data identify a new molecular mechanism of brain microvascular endothelial inflammatory response through NRP1-IFNγ crosstalk that could be a potential target for intervention of endothelial cell dysfunction in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ying Cao
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ashutosh K Mangalam
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa city, IA 52242, USA
| | - Yong Guo
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Jeffrey D Gamez
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Yuebo Zhang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Kirthica Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Baoan Ji
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Istvan Pirko
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Charles L Howe
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
24
|
Roth L, Prahst C, Ruckdeschel T, Savant S, Weström S, Fantin A, Riedel M, Héroult M, Ruhrberg C, Augustin HG. Neuropilin-1 mediates vascular permeability independently of vascular endothelial growth factor receptor-2 activation. Sci Signal 2016; 9:ra42. [PMID: 27117252 DOI: 10.1126/scisignal.aad3812] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neuropilin-1 (NRP1) regulates developmental and pathological angiogenesis, arteriogenesis, and vascular permeability, acting as a coreceptor for semaphorin 3A (Sema3A) and the 165-amino acid isoform of vascular endothelial growth factor A (VEGF-A165). NRP1 is also the receptor for the CendR peptides, a class of cell- and tissue-penetrating peptides with a specific R-x-x-R carboxyl-terminal motif. Because the cytoplasmic domain of NRP1 lacks catalytic activity, NRP1 is mainly thought to act through the recruitment and binding to other receptors. We report here that the NRP1 intracellular domain mediates vascular permeability. Stimulation with VEGF-A165, a ligand-blocking antibody, and a CendR peptide led to NRP1 accumulation at cell-cell contacts in endothelial cell monolayers, increased cellular permeability in vitro and vascular leakage in vivo. Biochemical analyses, VEGF receptor-2 (VEGFR-2) silencing, and the use of a specific VEGFR blocker established that the effects induced by the CendR peptide and the antibody were independent of VEGFR-2. Moreover, leakage assays in mice expressing a mutant NRP1 lacking the cytoplasmic domain revealed that this domain was required for NRP1-induced vascular permeability in vivo. Hence, these data define a vascular permeability pathway mediated by NRP1 but independent of VEGFR-2 activation.
Collapse
Affiliation(s)
- Lise Roth
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany. Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Claudia Prahst
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Tina Ruckdeschel
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Soniya Savant
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Simone Weström
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, EC1V 9EL London, UK
| | - Maria Riedel
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Mélanie Héroult
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, EC1V 9EL London, UK
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany. Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| |
Collapse
|
25
|
Raimondi C, Brash JT, Fantin A, Ruhrberg C. NRP1 function and targeting in neurovascular development and eye disease. Prog Retin Eye Res 2016; 52:64-83. [PMID: 26923176 PMCID: PMC4854174 DOI: 10.1016/j.preteyeres.2016.02.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 12/19/2022]
Abstract
Neuropilin 1 (NRP1) is expressed by neurons, blood vessels, immune cells and many other cell types in the mammalian body and binds a range of structurally and functionally diverse extracellular ligands to modulate organ development and function. In recent years, several types of mouse knockout models have been developed that have provided useful tools for experimental investigation of NRP1 function, and a multitude of therapeutics targeting NRP1 have been designed, mostly with the view to explore them for cancer treatment. This review provides a general overview of current knowledge of the signalling pathways that are modulated by NRP1, with particular focus on neuronal and vascular roles in the brain and retina. This review will also discuss the potential of NRP1 inhibitors for the treatment for neovascular eye diseases.
Collapse
Affiliation(s)
- Claudio Raimondi
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - James T Brash
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK.
| |
Collapse
|
26
|
Li X, Parker MW, Vander Kooi CW. Control of cellular motility by neuropilin-mediated physical interactions. Biomol Concepts 2015; 5:157-66. [PMID: 25018786 DOI: 10.1515/bmc-2013-0035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The neuropilin (Nrp) family consists of multifunctional cell surface receptors with critical roles in a number of different cell and tissue types. A core aspect of Nrp function is in ligand-dependent cellular migration, where it controls the multistep process of cellular motility through integration of ligand binding and receptor signaling. At a molecular level, the role of Nrp in migration is intimately connected to the control of adhesive interactions and cytoskeletal reorganization. Here, we review the physiological role of Nrp in cellular adhesion and motility in the cardiovascular and nervous systems. We also discuss the emerging pathological role of Nrp in tumor cell migration and metastasis, providing motivation for continued efforts toward developing Nrp inhibitors.
Collapse
|
27
|
Ellison TS, Atkinson SJ, Steri V, Kirkup BM, Preedy MEJ, Johnson RT, Ruhrberg C, Edwards DR, Schneider JG, Weilbaecher K, Robinson SD. Suppression of β3-integrin in mice triggers a neuropilin-1-dependent change in focal adhesion remodelling that can be targeted to block pathological angiogenesis. Dis Model Mech 2015; 8:1105-19. [PMID: 26159543 PMCID: PMC4582102 DOI: 10.1242/dmm.019927] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 07/07/2015] [Indexed: 12/12/2022] Open
Abstract
Anti-angiogenic treatments against αvβ3-integrin fail to block tumour growth in the long term, which suggests that the tumour vasculature escapes from angiogenesis inhibition through αvβ3-integrin-independent mechanisms. Here, we show that suppression of β3-integrin in mice leads to the activation of a neuropilin-1 (NRP1)-dependent cell migration pathway in endothelial cells via a mechanism that depends on NRP1's mobilisation away from mature focal adhesions following VEGF-stimulation. The simultaneous genetic targeting of both molecules significantly impairs paxillin-1 activation and focal adhesion remodelling in endothelial cells, and therefore inhibits tumour angiogenesis and the growth of already established tumours. These findings provide a firm foundation for testing drugs against these molecules in combination to treat patients with advanced cancers. Summary: Targeting both β3-integrin and neuropilin-1 prevents anti-angiogenic treatment escape.
Collapse
Affiliation(s)
- Tim S Ellison
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Samuel J Atkinson
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Veronica Steri
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Benjamin M Kirkup
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Michael E J Preedy
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Robert T Johnson
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | | | - Dylan R Edwards
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Jochen G Schneider
- Luxembourg Center for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg & Saarland University Medical Center, Internal Medicine II, L-4362 Homburg, Germany
| | - Katherine Weilbaecher
- Department of Internal Medicine, Division of Molecular Oncology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Stephen D Robinson
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| |
Collapse
|
28
|
Abstract
Neuropilins (NRPs) are highly conserved transmembrane glycoproteins that possess pleiotropic functions. Neuropilin-1 (NRP1) and its homologue neuropilin-2 interact as coreceptors with both class 3 semaphorins and vascular endothelial growth factor and are involved in neuronal guidance and angiogenesis, respectively. The contribution of NRPs to tumor angiogenesis has been highlighted in previous studies, leading to the development of NRP antagonists as novel anti-angiogenesis therapies. However, more recent studies have demonstrated that NRPs have a much broader spectrum of activity in the integration of different pathways in physiological and pathological conditions. A few studies investigated the role of NRPs in both malignant and non-neoplastic liver diseases. In normal liver, NRP1 is expressed in hepatic stellate cells and liver sinusoidal endothelial cells. NRP1 expression in hepatocytes has been associated with malignant transformation and may play an important role in tumor behavior. A contribution of NRPs in sinusoidal remodeling during liver regeneration has been also noted. Studies in chronic liver diseases have indicated that, besides its influence on angiogenesis, NRP1 might contribute to the progression of liver fibrosis owing to its effects on other growth factors, including transforming growth factor β1. As a result, NRP1 has been identified as a promising therapeutic target for future antifibrotic therapies based on the simultaneous blockade of multiple growth factor signaling pathways. In this review, the structure of NRPs and their interactions with various ligands and associated cell surface receptors are described briefly. The current understanding of the roles of the NRPs in liver diseases including tumors, regeneration and fibrogenesis, are also summarized.
Collapse
|
29
|
Bondeva T, Wolf G. Role of Neuropilin-1 in Diabetic Nephropathy. J Clin Med 2015; 4:1293-311. [PMID: 26239560 PMCID: PMC4485001 DOI: 10.3390/jcm4061293] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/28/2015] [Accepted: 06/09/2015] [Indexed: 02/07/2023] Open
Abstract
Diabetic nephropathy (DN) often develops in patients suffering from type 1 or type 2 diabetes mellitus. DN is characterized by renal injury resulting in proteinuria. Neuropilin-1 (NRP-1) is a single-pass transmembrane receptor protein devoid of enzymatic activity. Its large extracellular tail is structured in several domains, thereby allowing the molecule to interact with multiple ligands linking NRP-1 to different pathways through its signaling co-receptors. NRP-1’s role in nervous system development, immunity, and more recently in cancer, has been extensively investigated. Although its relation to regulation of apoptosis and cytoskeleton organization of glomerular vascular endothelial cells was reported, its function in diabetes mellitus and the development of DN is less clear. Several lines of evidence demonstrate a reduced NRP-1 expression in glycated-BSA cultured differentiated podocytes as well as in glomeruli from db/db mice (a model of type 2 Diabetes) and in diabetic patients diagnosed with DN. In vitro studies of podocytes implicated NRP-1 in the regulation of podocytes’ adhesion to extracellular matrix proteins, cytoskeleton reorganization, and apoptosis via not completely understood mechanisms. However, the exact role of NRP-1 during the onset of DN is not yet understood. This review intends to shed more light on NRP-1 and to present a link between NRP-1 and its signaling complexes in the development of DN.
Collapse
Affiliation(s)
- Tzvetanka Bondeva
- Department of Internal Medicine III, University Hospital Jena, Jena, 07747, Germany.
| | - Gunter Wolf
- Department of Internal Medicine III, University Hospital Jena, Jena, 07747, Germany.
| |
Collapse
|
30
|
Neuropilin-1 enforces extracellular matrix signalling via ABL1 to promote angiogenesis. Biochem Soc Trans 2015; 42:1429-34. [PMID: 25233427 DOI: 10.1042/bst20140141] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Neuropilin-1 (NRP1), together with neuropilin-2, belongs to the neuropilin family. Neuropilins are transmembrane proteins essential for vascular and neural development and act as co-receptors for secreted signalling molecules of the class 3 semaphorin and vascular endothelial growth factor A (VEGF-A) families. NRP1 promotes VEGF-A signal in blood vascular endothelium and semaphorin signal in lymphatic endothelium, by forming complexes with its co-receptors. Mouse mutant studies established that NRP1 expression is essential during development because mice lacking NRP1 expression die embryonically and show severe neuronal and cardiovascular defects. Even though the contribution of NRP1 to vascular development has been mainly ascribed to its function as a VEGF-A receptor, recent evidence suggests that NRP1 contributes to angiogenesis through VEGF-independent mechanisms. In the present paper, we provide an overview of NRP1 functions in the vasculature and discuss current knowledge of NRP1-dependent signalling in the endothelium.
Collapse
|
31
|
Ji T, Guo Y, Kim K, McQueen P, Ghaffar S, Christ A, Lin C, Eskander R, Zi X, Hoang BH. Neuropilin-2 expression is inhibited by secreted Wnt antagonists and its down-regulation is associated with reduced tumor growth and metastasis in osteosarcoma. Mol Cancer 2015; 14:86. [PMID: 25890345 PMCID: PMC4411772 DOI: 10.1186/s12943-015-0359-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 04/06/2015] [Indexed: 12/30/2022] Open
Abstract
Background Neuropilin 2 (NRP2) isa multi-functional co-receptor to many receptors, including VEGF receptor, c-Met and others. NRP2 has recently been implicated in tumor angiogenesis, growth, and metastasis of many other cancers. However, its role in osteosarcoma remains poorly understood. Results NRP2 was overexpressed in osteosarcoma cell lines and tissues, and associated with poor survival of osteosarcoma patients. Knockdown of NRP2 expression by short-hairpin (Sh) RNA resulted in reduced tumor growth, metastasis, and blood vessel formation of osteosarcoma. Knockdown of NRP2 expression by ShRNA also inhibited the recruitment of HUVEC cells to osteosarcoma cells. Inhibition of Wnt signaling by overexpression of secreted Wnt antagonists soluble LRP5, Frzb, and WIF1 markedly down-regulated mRNA and protein expression of NRP2 in osteosarcoma cell lines. Conclusions Regulation of NRP2 receptor expression may represent a novel approach for treatment of osteosarcoma through retarding osteosarcoma growth, metastasis and blood vessel formation. In addition, down-regulation of NRP2 expression can be achieved by expression of secreted Wnt antagonists. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0359-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tao Ji
- Department of Orthopaedic Surgery and Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA. .,Musculoskeletal Tumor Center, People's Hospital, Peking University, Beijing, China.
| | - Yi Guo
- Department of Orthopaedic Surgery and Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA.
| | - Kapjun Kim
- Department of Orthopaedic Surgery and Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA.
| | - Peter McQueen
- Department of Orthopaedic Surgery and Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA.
| | - Samia Ghaffar
- Department of Orthopaedic Surgery and Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA.
| | - Alexander Christ
- Department of Orthopaedic Surgery and Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA.
| | - Carol Lin
- Department of Oncology, CHOC Children's Hospital, Orange, CA, USA.
| | - Ramez Eskander
- Department of Obstetrics and Gynecology, University of California, Irvine, CA, USA.
| | - Xiaolin Zi
- Department of Urology and Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA.
| | - Bang H Hoang
- Department of Orthopaedic Surgery and Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, USA. .,Department of Orthopaedic Surgery, Montefiore Medical Center, The University Hospital for Albert Einstein College of Medicine, 3400 Bainbridge Ave, 6th Floor, Bronx, NY, 10476, USA.
| |
Collapse
|
32
|
Altered hippocampal-dependent memory and motor function in neuropilin 2-deficient mice. Transl Psychiatry 2015; 5:e521. [PMID: 25734514 PMCID: PMC4354347 DOI: 10.1038/tp.2015.17] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 12/16/2014] [Accepted: 01/12/2015] [Indexed: 01/19/2023] Open
Abstract
Semaphorins have an important role in synapse refinement in the mammalian nervous system. The class 3 semaphorin-3F (Sema3F) acting through neuropilin 2/plexin-A3 (Nrp2/PlexA3) holoreceptor complex signals in vivo to restrain apical dendritic spine morphogenesis of cortical pyramidal neurons and hippocampal neurons during postnatal development and mediates excitatory synaptic transmission. Semaphorin signaling has been implicated in the etiology of a number of neurodevelopmental disorders; however, the effects on behavior and mental function of dysregulated Sema3F-Nrp2 signaling have not been fully addressed. The present study is the first behavioral investigation of mice harboring a mutation of the nrp2 gene. Given that loss of Nrp2 signaling alters cortical and hippocampal synaptic organization, we investigated performance of nrp2-deficient mice on learning and sensorimotor function that are known to depend on cortical and hippocampal circuitry. When compared with age-matched controls, nrp2 null mice showed striking impairments in object recognition memory and preference for social novelty. In addition, nrp2(-/-) mice displayed impaired motor function in the rotarod test and in observations of grooming behavior. Exploration of novel olfactory sensory stimuli and nociception were unaffected by the loss of Nrp2. Overall, loss of Nrp2 may induce aberrant processing within hippocampal and corticostriatal networks that may contribute to neurodevelopmental disease mechanisms.
Collapse
|
33
|
Plein A, Fantin A, Ruhrberg C. Neuropilin regulation of angiogenesis, arteriogenesis, and vascular permeability. Microcirculation 2015; 21:315-23. [PMID: 24521511 PMCID: PMC4230468 DOI: 10.1111/micc.12124] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 02/05/2014] [Indexed: 12/11/2022]
Abstract
The formation of the cardiovasculature, consisting of both the heart and blood vessels, is a critical step in embryonic development and relies on three processes termed vasculogenesis, angiogenesis, and vascular remodeling. The transmembrane protein NRP1 is an essential modulator of embryonic angiogenesis with additional roles in vessel remodeling and arteriogenesis. NRP1 also enhances arteriogenesis in adults to alleviate pathological tissue ischemia. However, in certain circumstances, vascular NRP1 signaling can be detrimental, as it may promote cancer by enhancing tumor angiogenesis or contribute to tissue edema by increasing vascular permeability. Understanding the mechanisms of NRP1 signaling is, therefore, of profound importance for the design of therapies aiming to control vascular functions. Previous work has shown that vascular NRP1 can variably serve as a receptor for two secreted glycoproteins, the VEGF-A and SEMA3A, but it also has a poorly understood role as an adhesion receptor. Here, we review current knowledge of NRP1 function during blood vessel growth and homeostasis, with special emphasis on the vascular roles of its multiple ligands and signaling partners.
Collapse
Affiliation(s)
- Alice Plein
- UCL Institute of Ophthalmology, University College London, London, UK
| | | | | |
Collapse
|
34
|
Watanabe Y, Sakuma C, Yaginuma H. NRP1-mediated Sema3A signals coordinate laminar formation in the developing chick optic tectum. Development 2014; 141:3572-82. [PMID: 25183873 DOI: 10.1242/dev.110205] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The optic tectum comprises multiple layers, which are formed by radial and tangential migration during development. Here, we report that Neuropilin 1 (NRP1)-mediated Sema3A signals are involved in the process of tectal laminar formation, which is elaborated by tangential migration. In the developing chick tectum, NRP1, a receptor for Sema3A, is expressed in microtubule-associated protein 2 (MAP2)-positive intermediate layers IV and V. Sema3A itself is a diffusible guidance factor and is expressed in the overlying layer VI. Using stable fluorescent labeling of tectal cells, we show that MAP2-positive intermediate layers are formed by the neurons that have been dispersed by tangential migration along the tectal efferent axons. When Sema3A was mis-expressed during laminar formation, local Sema3A repelled the tangential migrants, thus eliminating MAP2-positive neurons that expressed NRP1. Furthermore, in the absence of the MAP2-positive neurons, tectal layers were disorganized into an undulated form, indicating that MAP2-positive intermediate layers are required for proper laminar formation. These results suggest that NRP1-mediated Sema3A signals provide repulsive signals for MAP2-positive neurons to segregate tectal layers, which is important in order to coordinate laminar organization of the optic tectum.
Collapse
Affiliation(s)
- Yuji Watanabe
- Department of Molecular Neurobiology, Graduate School of Life Sciences, Tohoku University, Aoba-ku, Sendai 980-8575, Japan Institute of Development, Aging & Cancer, Tohoku University, Sendai 980-8575, Japan Department of Neuroanatomy and Embryology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Chie Sakuma
- Department of Neuroanatomy and Embryology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hiroyuki Yaginuma
- Department of Neuroanatomy and Embryology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| |
Collapse
|
35
|
A role for neuropilins in the interaction between Schwann cells and meningeal cells. PLoS One 2014; 9:e109401. [PMID: 25314276 PMCID: PMC4196904 DOI: 10.1371/journal.pone.0109401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 09/10/2014] [Indexed: 11/28/2022] Open
Abstract
In their natural habitat, the peripheral nerve, Schwann cells (SCs) form nicely aligned pathways (also known as the bands of Büngner) that guide regenerating axons to their targets. Schwann cells that are implanted in the lesioned spinal cord fail to align in pathways that could support axon growth but form cellular clusters that exhibit only limited intermingling with the astrocytes and meningeal cells (MCs) that are present in the neural scar. The formation of cell clusters can be studied in co-cultures of SCs and MCs. In these co-cultures SCs form cluster-like non-overlapping cell aggregates with well-defined boundaries. There are several indications that neuropilins (NRPs) play an important role in MC-induced SC aggregation. Both SCs and MCs express NRP1 and NRP2 and SCs express the NRP ligands Sema3B, C and E while MCs express Sema3A, C, E and F. We now demonstrate that in SC-MC co-cultures, siRNA mediated knockdown of NRP2 in SCs decreased the formation of SC clusters while these SCs maintained their capacity to align in bands of Büngner-like columnar arrays. Unexpectedly, knockdown of NRP1 expression resulted in a significant increase in SC aggregation. These results suggest that a reduction in NRP2 expression may enhance the capacity of implanted SCs to interact with MCs that invade a neural scar formed after a lesion of the spinal cord.
Collapse
|
36
|
ADAM metalloproteases promote a developmental switch in responsiveness to the axonal repellant Sema3A. Nat Commun 2014; 5:4058. [PMID: 24898499 DOI: 10.1038/ncomms5058] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/06/2014] [Indexed: 11/08/2022] Open
Abstract
During embryonic development, axons can gain and lose sensitivity to guidance cues, and this flexibility is essential for the correct wiring of the nervous system. Yet, the underlying molecular mechanisms are largely unknown. Here we show that receptor cleavage by ADAM (A Disintegrin And Metalloprotease) metalloproteases promotes murine sensory axons loss of responsiveness to the chemorepellant Sema3A. Genetic ablation of ADAM10 and ADAM17 disrupts the developmental downregulation of Neuropilin-1 (Nrp1), the receptor for Sema3A, in sensory axons. Moreover, this is correlated with gain of repulsive response to Sema3A. Overexpression of Nrp1 in neurons reverses axonal desensitization to Sema3A, but this is hampered in a mutant Nrp1 with high susceptibility to cleavage. Lastly, we detect guidance errors of proprioceptive axons in ADAM knockouts that are consistent with enhanced response to Sema3A. Our results provide the first evidence for involvement of ADAMs in regulating developmental switch in responsiveness to axonal guidance cues.
Collapse
|
37
|
Roet KCD, Verhaagen J. Understanding the neural repair-promoting properties of olfactory ensheathing cells. Exp Neurol 2014; 261:594-609. [PMID: 24842489 DOI: 10.1016/j.expneurol.2014.05.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 12/13/2022]
Abstract
Olfactory ensheathing glial cells (OECs) are a specialized type of glia that form a continuously aligned cellular pathway that actively supports unprecedented regeneration of primary olfactory axons from the periphery into the central nervous system. Implantation of OECs stimulates neural repair in experimental models of spinal cord, brain and peripheral nerve injury and delays disease progression in animal models for neurodegenerative diseases like amyotrophic lateral sclerosis. OECs implanted in the injured spinal cord display a plethora of pro-regenerative effects; they promote axonal regeneration, reorganize the glial scar, remyelinate axons, stimulate blood vessel formation, have phagocytic properties and modulate the immune response. Recently genome wide transcriptional profiling and proteomics analysis combined with classical or larger scale "medium-throughput" bioassays have provided novel insights into the molecular mechanism that endow OECs with their pro-regenerative properties. Here we review these studies and show that the gaps that existed in our understanding of the molecular basis of the reparative properties of OECs are narrowing. OECs express functionally connected sets of genes that can be linked to at least 10 distinct processes directly relevant to neural repair. The data indicate that OECs exhibit a range of synergistic cellular activities, including active and passive stimulation of axon regeneration (by secretion of growth factors, axon guidance molecules and basement membrane components) and critical aspects of tissue repair (by structural remodeling and support, modulation of the immune system, enhancement of neurotrophic and antigenic stimuli and by metabolizing toxic macromolecules). Future experimentation will have to further explore the newly acquired knowledge to enhance the therapeutic potential of OECs.
Collapse
Affiliation(s)
- Kasper C D Roet
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105BA Amsterdam, The Netherlands.
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105BA Amsterdam, The Netherlands; Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Boelelaan 1085, Amsterdam 1081HV, The Netherlands.
| |
Collapse
|
38
|
Koch S, van Meeteren LA, Morin E, Testini C, Weström S, Björkelund H, Le Jan S, Adler J, Berger P, Claesson-Welsh L. NRP1 presented in trans to the endothelium arrests VEGFR2 endocytosis, preventing angiogenic signaling and tumor initiation. Dev Cell 2014; 28:633-46. [PMID: 24656741 DOI: 10.1016/j.devcel.2014.02.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 11/04/2013] [Accepted: 02/13/2014] [Indexed: 11/30/2022]
Abstract
Neuropilin 1 (NRP1) modulates angiogenesis by binding vascular endothelial growth factor (VEGF) and its receptor, VEGFR2. We examined the consequences when VEGFR2 and NRP1 were expressed on the same cell (cis) or on different cells (trans). In cis, VEGF induced rapid VEGFR2/NRP1 complex formation and internalization. In trans, complex formation was delayed and phosphorylation of phospholipase Cγ (PLCγ) and extracellular regulated kinase 2 (ERK2) was prolonged, whereas ERK1 phosphorylation was reduced. Trans complex formation suppressed initiation and vascularization of NRP1-expressing mouse fibrosarcoma and melanoma. Suppression in trans required high-affinity, steady-state binding of VEGF to NRP1, which was dependent on the NRP1 C-terminal domain. Compatible with a trans effect of NRP1, quiescent vasculature in the developing retina showed continuous high NRP1 expression, whereas angiogenic sprouting occurred where NRP1 levels fluctuated between adjacent endothelial cells. Therefore, through communication in trans, NRP1 can modulate VEGFR2 signaling and suppress angiogenesis.
Collapse
Affiliation(s)
- Sina Koch
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory and Science for Life Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 75185 Uppsala, Sweden
| | - Laurens A van Meeteren
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory and Science for Life Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 75185 Uppsala, Sweden
| | - Eric Morin
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory and Science for Life Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 75185 Uppsala, Sweden
| | - Chiara Testini
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory and Science for Life Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 75185 Uppsala, Sweden
| | - Simone Weström
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory and Science for Life Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 75185 Uppsala, Sweden
| | | | - Sébastien Le Jan
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory and Science for Life Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 75185 Uppsala, Sweden
| | - Jeremy Adler
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory and Science for Life Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 75185 Uppsala, Sweden
| | - Philipp Berger
- Paul Scherrer Institute, Laboratory of Biomolecular Research, Molecular Cell Biology, 5232 Villigen PSI, Switzerland
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory and Science for Life Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 75185 Uppsala, Sweden.
| |
Collapse
|
39
|
Roet KCD, Franssen EHP, de Bree FM, Essing AHW, Zijlstra SJJ, Fagoe ND, Eggink HM, Eggers R, Smit AB, van Kesteren RE, Verhaagen J. A multilevel screening strategy defines a molecular fingerprint of proregenerative olfactory ensheathing cells and identifies SCARB2, a protein that improves regenerative sprouting of injured sensory spinal axons. J Neurosci 2013; 33:11116-35. [PMID: 23825416 PMCID: PMC6618611 DOI: 10.1523/jneurosci.1002-13.2013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 05/14/2013] [Accepted: 05/15/2013] [Indexed: 11/21/2022] Open
Abstract
Olfactory ensheathing cells (OECs) have neuro-restorative properties in animal models for spinal cord injury, stroke, and amyotrophic lateral sclerosis. Here we used a multistep screening approach to discover genes specifically contributing to the regeneration-promoting properties of OECs. Microarray screening of the injured olfactory pathway and of cultured OECs identified 102 genes that were subsequently functionally characterized in cocultures of OECs and primary dorsal root ganglion (DRG) neurons. Selective siRNA-mediated knockdown of 16 genes in OECs (ADAMTS1, BM385941, FZD1, GFRA1, LEPRE1, NCAM1, NID2, NRP1, MSLN, RND1, S100A9, SCARB2, SERPINI1, SERPINF1, TGFB2, and VAV1) significantly reduced outgrowth of cocultured DRG neurons, indicating that endogenous expression of these genes in OECs supports neurite extension of DRG neurons. In a gain-of-function screen for 18 genes, six (CX3CL1, FZD1, LEPRE1, S100A9, SCARB2, and SERPINI1) enhanced and one (TIMP2) inhibited neurite growth. The most potent hit in both the loss- and gain-of-function screens was SCARB2, a protein that promotes cholesterol secretion. Transplants of fibroblasts that were genetically modified to overexpress SCARB2 significantly increased the number of regenerating DRG axons that grew toward the center of a spinal cord lesion in rats. We conclude that expression of SCARB2 enhances regenerative sprouting and that SCARB2 contributes to OEC-mediated neuronal repair.
Collapse
Affiliation(s)
- Kasper C D Roet
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Cao Y, Hoeppner LH, Bach S, E G, Guo Y, Wang E, Wu J, Cowley MJ, Chang DK, Waddell N, Grimmond SM, Biankin AV, Daly RJ, Zhang X, Mukhopadhyay D. Neuropilin-2 promotes extravasation and metastasis by interacting with endothelial α5 integrin. Cancer Res 2013; 73:4579-4590. [PMID: 23689123 DOI: 10.1158/0008-5472.can-13-0529] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Metastasis, the leading cause of cancer death, requires tumor cell intravasation, migration through the bloodstream, arrest within capillaries, and extravasation to invade distant tissues. Few mechanistic details have been reported thus far regarding the extravasation process or re-entry of circulating tumor cells at metastatic sites. Here, we show that neuropilin-2 (NRP-2), a multifunctional nonkinase receptor for semaphorins, vascular endothelial growth factor (VEGF), and other growth factors, expressed on cancer cells interacts with α5 integrin on endothelial cells to mediate vascular extravasation and metastasis in zebrafish and murine xenograft models of clear cell renal cell carcinoma (RCC) and pancreatic adenocarcinoma. In tissue from patients with RCC, NRP-2 expression is positively correlated with tumor grade and is highest in metastatic tumors. In a prospectively acquired cohort of patients with pancreatic cancer, high NRP-2 expression cosegregated with poor prognosis. Through biochemical approaches as well as Atomic Force Microscopy (AFM), we describe a unique mechanism through which NRP-2 expressed on cancer cells interacts with α5 integrin on endothelial cells to mediate vascular adhesion and extravasation. Taken together, our studies reveal a clinically significant role of NRP-2 in cancer cell extravasation and promotion of metastasis.
Collapse
Affiliation(s)
- Ying Cao
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Luke H Hoeppner
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Steven Bach
- Bioengineering Program & Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, PA 18015
| | - Guangqi E
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Yan Guo
- Bioengineering Program & Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, PA 18015
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Jianmin Wu
- The Kinghorn Cancer Centre, Cancer Research Division, Garvan Institute of Medical Research, 370 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
| | - Mark J Cowley
- The Kinghorn Cancer Centre, Cancer Research Division, Garvan Institute of Medical Research, 370 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
| | - David K Chang
- The Kinghorn Cancer Centre, Cancer Research Division, Garvan Institute of Medical Research, 370 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.,Department of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, NSW 2200, Australia.,South Western Sydney Clinical School, Faculty of Medicine, University of NSW, Liverpool NSW 2170, Australia
| | - Nicola Waddell
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Sean M Grimmond
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Andrew V Biankin
- The Kinghorn Cancer Centre, Cancer Research Division, Garvan Institute of Medical Research, 370 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.,Department of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, NSW 2200, Australia.,South Western Sydney Clinical School, Faculty of Medicine, University of NSW, Liverpool NSW 2170, Australia
| | - Roger J Daly
- The Kinghorn Cancer Centre, Cancer Research Division, Garvan Institute of Medical Research, 370 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
| | - Xiaohui Zhang
- Bioengineering Program & Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, PA 18015
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
41
|
Torigoe M, Yamauchi K, Tamada A, Matsuda I, Aiba A, Castellani V, Murakami F. Role of neuropilin-2 in the ipsilateral growth of midbrain dopaminergic axons. Eur J Neurosci 2013; 37:1573-83. [PMID: 23534961 DOI: 10.1111/ejn.12190] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 02/13/2013] [Accepted: 02/14/2013] [Indexed: 01/01/2023]
Abstract
Axonal projections in the CNS can be categorized as either crossed or uncrossed. Crossing and uncrossing of axons has been explained by attractive and repulsive molecules like Netrin-1 and Slits, which are secreted by midline structures. However, uncrossed projections can be established even in double knockout mice of slit1 and slit2 or of roundabout1 (robo1) and robo2, two receptors for Slits. Here, we found that a novel mechanism mediated by Neuropilin-2 (Nrp2) contributes to the formation of uncrossed projections of midbrain dopaminergic neurons (mDANs). Nrp2 transcriptional activities were detected in a subset of mDANs, and its protein was expressed in mDAN axons growing through the ipsilateral diencephalon. In nrp2(lac) (Z) (/lac) (Z) mice, mDAN axons aberrantly grew toward the ventral midline and even crossed it, suggesting that Nrp2 is necessary for the development of mDAN ipsilateral projections. We investigated the involvement of Semaphorin 3B (Sema3B) and Sema3F, two ligands of Nrp2, by analysing mDAN axon trajectories in single or double knockout mice. In both cases, mDAN axons still projected ipsilaterally, suggesting the involvement mechanisms independent of these Sema3s. Nrp2-deficient mDAN axons retained their responsiveness to Slit2, demonstrating that aberrant mDAN axons in nrp2(lac) (Z) (/lac) (Z) mice were not indirectly mediated by alterations in Slit/Robo signaling. Taken together, our results indicate that a novel mechanism mediated by Nrp2 contributes to the establishment of uncrossed projections by mDAN axons.
Collapse
Affiliation(s)
- Makio Torigoe
- Laboratory of Neuroscience, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
NRP1 acts cell autonomously in endothelium to promote tip cell function during sprouting angiogenesis. Blood 2013; 121:2352-62. [PMID: 23315162 DOI: 10.1182/blood-2012-05-424713] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Neuropilin (NRP) 1 is a receptor for the vascular endothelial growth factor (VEGF)-A and is essential for normal angiogenesis. Previous in vitro experiments identified NRP1 interactions with VEGF-A's main signaling receptor VEGFR2 within endothelial cells, but also between nonendothelial NRP1 and endothelial VEGFR2. Consistent with an endothelial role for NRP1 in angiogenesis, we found that VEGFR2 and NRP1 were coexpressed in endothelial tip and stalk cells in the developing brain. In addition, NRP1 was expressed on two cell types that interact with growing brain vessels-the neural progenitors that secrete VEGF-A to stimulate tip cell activity and the pro-angiogenic macrophages that promote tip cell anastomosis. Selective targeting of Nrp1 in each of these cell types demonstrated that neural progenitor- and macrophage-derived NRP1 were dispensable, whereas endothelial NRP1 was essential for normal brain vessel growth. NRP1 therefore promotes brain angiogenesis cell autonomously in endothelium, independently of heterotypic interactions with nonendothelial cells. Genetic mosaic analyses demonstrated a key role for NRP1 in endothelial tip rather than stalk cells during vessel sprouting. Thus, NRP1-expressing endothelial cells attained the tip cell position when competing with NRP1-negative endothelial cells in chimeric vessel sprouts. Taken together, these findings demonstrate that NRP1 promotes endothelial tip cell function during angiogenesis.
Collapse
|
43
|
Parker MW, Guo HF, Li X, Linkugel AD, Vander Kooi CW. Function of members of the neuropilin family as essential pleiotropic cell surface receptors. Biochemistry 2012; 51:9437-46. [PMID: 23116416 DOI: 10.1021/bi3012143] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The neuropilin (Nrp) family consists of essential multifunctional vertebrate cell surface receptors. Nrps were initially characterized as receptors for class III Semaphorin (Sema3) family members, functioning in axon guidance. Nrps have also been shown to be critical for vascular endothelial growth factor-dependent angiogenesis. Intriguingly, recent data show that Nrp function in these seemingly divergent pathways is critically determined by ligand-mediated cross-talk, which underlies Nrp function in both physiological and pathological processes. In addition to functioning in these two pathways, Nrps have been shown to specifically function in a number of other fundamental signaling pathways as well. Multiple general mechanisms have been found to directly contribute to the pleiotropic function of Nrp. Here we review critical general features of Nrps that function as essential receptors integrating multiple molecular cues into diverse cellular signaling.
Collapse
Affiliation(s)
- Matthew W Parker
- Department of Molecular and Cellular Biochemistry, Center for Structural Biology, University of Kentucky, Lexington, KY 40536, USA
| | | | | | | | | |
Collapse
|
44
|
Sijaona A, Luukko K, Kvinnsland IH, Kettunen P. Expression patterns of Sema3F, PlexinA4, -A3, Neuropilin1 and -2 in the postnatal mouse molar suggest roles in tooth innervation and organogenesis. Acta Odontol Scand 2012; 70:140-8. [PMID: 21815834 DOI: 10.3109/00016357.2011.600708] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Semaphorins form a family of axon wiring molecules but still little is known about their role in tooth formation. A class 3 semaphorin, Semaphorin3F (Sema3F), besides acting as a chemorepellant for different types of axons, controls a variety of non-neuronal developmental processes. MATERIALS AND METHODS Cellular mRNA expression patterns of Sema3F as well as neuropilin 1 (Npn1), neuropilin 2 (Npn2), plexinA3 and plexinA4 receptors were analyzed by sectional in situ hybridization in the mouse molar tooth during postnatal days 0-7. The expression of the receptors was studied in PN5 trigeminal ganglia. RESULTS Sema3F, Npn1, -2 and plexinA4 exhibited distinct, spatiotemporally changing expression patterns, whereas plexinA3 was not observed in the tooth germs. Besides being expressed in the base of the dental mesenchyme Sema3F, like plexinA4, Npn1 and -2, was present in the ameloblast cell lineage. Npn1 and Npn2 were additionally seen in the pulp horns and endothelial cells and like PlexinA4 in the developing alveolar bone. Npn1, plexinA3 and -A4 were observed in trigeminal ganglion neurons. CONCLUSIONS Sema3F may act as a tooth target-derived axonal chemorepellant controlling establishment of the tooth nerve supply. Furthermore, Sema3F, like Npn1, -2 and plexinA4 may serve non-neuronal functions by controlling the development of the ameloblast cell lineage. Moreover, Npn1 and Npn2 may regulate dental vasculogenesis and, together with PlexinA4, alveolar bone formation. Further analyses such as investigation of transgenic mouse models will be required to elucidate in vivo signaling functions of Sema3F and the receptors in odontogenesis.
Collapse
|
45
|
Hillman RT, Feng BY, Ni J, Woo WM, Milenkovic L, Hayden Gephart MG, Teruel MN, Oro AE, Chen JK, Scott MP. Neuropilins are positive regulators of Hedgehog signal transduction. Genes Dev 2011; 25:2333-46. [PMID: 22051878 DOI: 10.1101/gad.173054.111] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Hedgehog (Hh) pathway is essential for vertebrate embryogenesis, and excessive Hh target gene activation can cause cancer in humans. Here we show that Neuropilin 1 (Nrp1) and Nrp2, transmembrane proteins with roles in axon guidance and vascular endothelial growth factor (VEGF) signaling, are important positive regulators of Hh signal transduction. Nrps are expressed at times and locations of active Hh signal transduction during mouse development. Using cell lines lacking key Hh pathway components, we show that Nrps mediate Hh transduction between activated Smoothened (Smo) protein and the negative regulator Suppressor of Fused (SuFu). Nrp1 transcription is induced by Hh signaling, and Nrp1 overexpression increases maximal Hh target gene activation, indicating the existence of a positive feedback circuit. The regulation of Hh signal transduction by Nrps is conserved between mammals and bony fish, as we show that morpholinos targeting the Nrp zebrafish ortholog nrp1a produce a specific and highly penetrant Hh pathway loss-of-function phenotype. These findings enhance our knowledge of Hh pathway regulation and provide evidence for a conserved nexus between Nrps and this important developmental signaling system.
Collapse
Affiliation(s)
- R Tyler Hillman
- Department of Genetics, Stanford University School of Medicine, California 94305, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Pereira Lopes FR, Lisboa BCG, Frattini F, Almeida FM, Tomaz MA, Matsumoto PK, Langone F, Lora S, Melo PA, Borojevic R, Han SW, Martinez AMB. Enhancement of sciatic nerve regeneration after vascular endothelial growth factor (VEGF) gene therapy. Neuropathol Appl Neurobiol 2011; 37:600-12. [DOI: 10.1111/j.1365-2990.2011.01159.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
47
|
Nawabi H, Castellani V. Axonal commissures in the central nervous system: how to cross the midline? Cell Mol Life Sci 2011; 68:2539-53. [PMID: 21538161 PMCID: PMC11114790 DOI: 10.1007/s00018-011-0691-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 04/08/2011] [Accepted: 04/14/2011] [Indexed: 01/02/2023]
Abstract
Organisms with bilateral symmetry elaborate patterns of neuronal projections connecting both sides of the central nervous system at all levels of the neuraxis. During development, these so-called commissural projections navigate across the midline to innervate their contralateral targets. Commissural axon pathfinding has been extensively studied over the past years and turns out to be a highly complex process, implicating modulation of axon responsiveness to the various guidance cues that instruct axon trajectories towards, within and away from the midline. Understanding the molecular mechanisms allowing these switches of response to take place at the appropriate time and place is a major challenge for current research. Recent work characterized several instructive processes controlling the spatial and temporal fine-tuning of the guidance molecular machinery. These findings illustrate the molecular strategies by which commissural axons modulate their sensitivity to guidance cues during midline crossing and show that regulation at both transcriptional and post-transcriptional levels are crucial for commissural axon guidance.
Collapse
Affiliation(s)
- Homaira Nawabi
- F.M. Kirby Neurobiology Center, Children's Hospital and Department of Neurology, Harvard Medical School, Boston, MA 02115, USA.
| | | |
Collapse
|
48
|
Sullivan LA, Brekken RA. The VEGF family in cancer and antibody-based strategies for their inhibition. MAbs 2011; 2:165-75. [PMID: 20190566 DOI: 10.4161/mabs.2.2.11360] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is required in normal physiological processes, but is also involved in tumor growth, progression and metastasis. Vascular endothelial growth factor (VEGF), a primary mediator of angiogenesis in normal physiology and in disease, and other VEGF family members and their receptors provide targets that have been explored extensively for cancer therapy. Small molecule inhibitors and antibody/protein-based strategies that target the VEGF pathway have been studied in multiple types of cancer. This review will focus on VEGF pathway targeting antibodies that are currently being evaluated in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Laura A Sullivan
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, USA
| | | |
Collapse
|
49
|
Grandclement C, Borg C. Neuropilins: a new target for cancer therapy. Cancers (Basel) 2011; 3:1899-928. [PMID: 24212788 PMCID: PMC3757396 DOI: 10.3390/cancers3021899] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 03/23/2011] [Accepted: 04/01/2011] [Indexed: 02/07/2023] Open
Abstract
Recent investigations highlighted strong similarities between neural crest migration during embryogenesis and metastatic processes. Indeed, some families of axon guidance molecules were also reported to participate in cancer invasion: plexins/semaphorins/neuropilins, ephrins/Eph receptors, netrin/DCC/UNC5. Neuropilins (NRPs) are transmembrane non tyrosine-kinase glycoproteins first identified as receptors for class-3 semaphorins. They are particularly involved in neural crest migration and axonal growth during development of the nervous system. Since many types of tumor and endothelial cells express NRP receptors, various soluble molecules were also found to interact with these receptors to modulate cancer progression. Among them, angiogenic factors belonging to the Vascular Endothelial Growth Factor (VEGF) family seem to be responsible for NRP-related angiogenesis. Because NRPs expression is often upregulated in cancer tissues and correlated with poor prognosis, NRPs expression might be considered as a prognostic factor. While NRP1 was intensively studied for many years and identified as an attractive angiogenesis target for cancer therapy, the NRP2 signaling pathway has just recently been studied. Although NRP genes share 44% homology, differences in their expression patterns, ligands specificities and signaling pathways were observed. Indeed, NRP2 may regulate tumor progression by several concurrent mechanisms, not only angiogenesis but lymphangiogenesis, epithelial-mesenchymal transition and metastasis. In view of their multiples functions in cancer promotion, NRPs fulfill all the criteria of a therapeutic target for innovative anti-tumor therapies. This review focuses on NRP-specific roles in tumor progression.
Collapse
Affiliation(s)
- Camille Grandclement
- INSERM UMR 645, F-25020 Besançon, France; E-Mail:
- University of Franche-Comté, IFR133, F-25020 Besançon, France
- EFS Bourgogne Franche-Comté, F-25020 Besançon, France
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +33-3-81-61-56-15 or +33-3-81-66-93-21; Fax: +33-3-81-61-56-17
| | - Christophe Borg
- INSERM UMR 645, F-25020 Besançon, France; E-Mail:
- University of Franche-Comté, IFR133, F-25020 Besançon, France
- EFS Bourgogne Franche-Comté, F-25020 Besançon, France
- Department of Medical Oncology, CHU Besançon, F-25000 Besançon, France
| |
Collapse
|
50
|
Abstract
Virtually all metazoan cells contain at least one and usually several types of transmembrane proteoglycans. These are varied in protein structure and type of polysaccharide, but the total number of vertebrate genes encoding transmembrane proteoglycan core proteins is less than 10. Some core proteins, including those of the syndecans, always possess covalently coupled glycosaminoglycans; others do not. Syndecan has a long evolutionary history, as it is present in invertebrates, but many other transmembrane proteoglycans are vertebrate inventions. The variety of proteins and their glycosaminoglycan chains is matched by diverse functions. However, all assume roles as coreceptors, often working alongside high-affinity growth factor receptors or adhesion receptors such as integrins. Other common themes are an ability to signal through their cytoplasmic domains, often to the actin cytoskeleton, and linkage to PDZ protein networks. Many transmembrane proteoglycans associate on the cell surface with metzincin proteases and can be shed by them. Work with model systems in vivo and in vitro reveals roles in growth, adhesion, migration, and metabolism. Furthermore, a wide range of phenotypes for the core proteins has been obtained in mouse knockout experiments. Here some of the latest developments in the field are examined in hopes of stimulating further interest in this fascinating group of molecules.
Collapse
Affiliation(s)
- John R Couchman
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, 2200 Denmark.
| |
Collapse
|