1
|
Lee C, Kim MJ, Kumar A, Lee HW, Yang Y, Kim Y. Vascular endothelial growth factor signaling in health and disease: from molecular mechanisms to therapeutic perspectives. Signal Transduct Target Ther 2025; 10:170. [PMID: 40383803 PMCID: PMC12086256 DOI: 10.1038/s41392-025-02249-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/09/2025] [Accepted: 04/21/2025] [Indexed: 05/20/2025] Open
Abstract
Vascular endothelial growth factor (VEGF) signaling is a critical regulator of vasculogenesis, angiogenesis, and lymphangiogenesis, processes that are vital for the development of vascular and lymphatic systems, tissue repair, and the maintenance of homeostasis. VEGF ligands and their receptors orchestrate endothelial cell proliferation, migration, and survival, playing a pivotal role in dynamic vascular remodeling. Dysregulated VEGF signaling drives diverse pathological conditions, including tumor angiogenesis, cardiovascular diseases, and ocular disorders. Excessive VEGF activity promotes tumor growth, invasion, and metastasis, while insufficient signaling contributes to impaired wound healing and ischemic diseases. VEGF-targeted therapies, such as monoclonal antibodies and tyrosine kinase inhibitors, have revolutionized the treatment of diseases involving pathological angiogenesis, offering significant clinical benefits in oncology and ophthalmology. These therapies inhibit angiogenesis and slow disease progression, but they often face challenges such as therapeutic resistance, suboptimal efficacy, and adverse effects. To further explore these issues, this review provides a comprehensive overview of VEGF ligands and receptors, elucidating their molecular mechanisms and regulatory networks. It evaluates the latest progress in VEGF-targeted therapies and examines strategies to address current challenges, such as resistance mechanisms. Moreover, the discussion includes emerging therapeutic strategies such as innovative drug delivery systems and combination therapies, highlighting the continuous efforts to improve the effectiveness and safety of VEGF-targeted treatments. This review highlights the translational potential of recent discoveries in VEGF biology for improving patient outcomes.
Collapse
Affiliation(s)
- Chunsik Lee
- Department of R&D, GEMCRO Inc, Seoul, Republic of Korea.
| | - Myung-Jin Kim
- Department of Biological Sciences and Research Institute of Women's Health, Sookmyung Women's University, Seoul, Republic of Korea
| | - Anil Kumar
- Center for Research and Innovations, Adichunchanagiri University, Mandya, Karnataka, India
| | - Han-Woong Lee
- Department of R&D, GEMCRO Inc, Seoul, Republic of Korea
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yonghwan Kim
- Department of Biological Sciences and Research Institute of Women's Health, Sookmyung Women's University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Li C, Cui X, Ren M, Yin Y, He S. Identification of biomarkers and potential drug targets for esophageal cancer: a Mendelian randomization study. Sci Rep 2025; 15:8176. [PMID: 40059241 PMCID: PMC11891310 DOI: 10.1038/s41598-025-93068-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/04/2025] [Indexed: 05/13/2025] Open
Abstract
Esophageal cancer (EC) is a common and deadly malignancy of the digestive system. Currently, effective treatments for EC are limited and patient prognosis remains poor. In this study, we utilized Mendelian Randomization (MR) to identify potential drug targets for EC by analyzing proteins linked to the disease risk. A total of 734 plasma proteins and 4,479 druggable genes were obtained from recent studies, and two-sample MR analyses were conducted to investigate causal relationships between these proteins and EC. The cis-pQTL data of the proteins was analyzed after filtering. The inverse variance weighted (IVW) method was the primary analytical approach in MR analysis. Steiger filtering, heterogeneity and pleiotropy tests, Summary-data-based Mendelian Randomization (SMR) analysis, and Bayesian co-localization analysis were implemented to consolidate the results further. Moreover, drugs corresponding to the identified proteins were found in the DrugBank database. Five proteins HPSE, ST3GAL1, CEL, KLK13, and GNRH2 were identified as highly associated with EC. HPSE and GNRH2 showed protective effects with odds ratios (OR) of 0.80 (95% confidence interval [CI], 0.70-0.92) and 0.73 (95% CI 0.54-0.98), respectively. In contrast, increased expression of ST3GAL1(OR, 1.37; 95% CI 1.04-1.82), CEL (OR, 1.27; 95% CI 1.08-1.49), and KLK13 (OR, 1.22; 95% CI 1.04-1.42) were all associated with a higher risk of EC. In addition, the HPSE protein showed moderate colocalization with EC [coloc.abf-posterior probability of hypothesis 4 (PPH4) = 0.637]. Furthermore, the sensitivity analyses indicated no heterogeneity or pleiotropy. Therefore, these findings present promising drug targets for EC and deserve further clinical investigation.
Collapse
Affiliation(s)
- Chengjun Li
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xiaomeng Cui
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Mudan Ren
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yan Yin
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Shuixiang He
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
3
|
Erning K, Wilson KL, Smith CS, Nguyen L, Joesph NI, Irengo R, Cao LY, Cumaran M, Shi Y, Lyu S, Riley L, Dunn TW, Carmichael ST, Segura T. Clustered VEGF Nanoparticles in Microporous Annealed Particle (MAP) Hydrogel Accelerates Functional Recovery and Brain Tissue Repair after Stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635733. [PMID: 39974959 PMCID: PMC11838428 DOI: 10.1101/2025.01.30.635733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Ischemic stroke, a blockage in the vasculature of the brain that results in insufficient blood flow, is one of the world's leading causes of disability. The cascade of inflammation and cell death that occurs immediately following stroke drives vascular and functional loss that does not fully recover over time, and no FDA-approved therapies exist that stimulate regeneration post-stroke. We have previously developed a hydrogel scaffold that delivered heparin nanoparticles with and without VEGF bound to their surface to promote angiogenesis and reduce inflammation, respectively. However, the inclusion of the naked heparin nanoparticles warranted concern over the development of bleeding complications. Here, we explore how microporous annealed particle (MAP) scaffolds functionalized with VEGF coated heparin nanoparticles can both reduce inflammation and promote angiogenesis - without the inclusion of free heparin nanoparticles. We show that our updated design not only successfully promotes de novo tissue formation, including the development of mature vessels and neurite sprouting, but it also leads to functional improvement in a photothrombotic stroke model. In addition, we find increased astrocyte infiltration into the infarct site correlated with mature vessel formation. This work demonstrates how our biomaterial design can enhance endogenous regeneration post-stroke while eliminating the need for excess heparin.
Collapse
|
4
|
Fathi Z, Boojar MMA, Sajedi RH, Dehnavi E, Jahanafrooz Z. Expression of VEGFR2 Ligand Binding Domain in Pichia pink™ 4 Cells and Evaluation of Its Interactions with VEGF-A 165 Receptor Binding Domain. Mol Biotechnol 2025; 67:342-355. [PMID: 38349458 DOI: 10.1007/s12033-024-01057-1] [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: 09/07/2023] [Accepted: 12/26/2023] [Indexed: 01/04/2025]
Abstract
Vascular endothelial growth factor A165 (VEGF-A165) and VEGF receptor 2 (KDR) are important mediators of angiogenesis. We aimed to express the soluble KDR ligand-binding domain (sKDR1-3) and evaluate its interaction with the VEGF-A165 receptor-binding domain (VEGFA165-RBD). sKDR1-3 DNA was designed and subcloned into pPinkα-HC plasmid. The cassette was transfected into the Pichia pink™ 4 genome by homologous recombination. We optimized the expression of sKDR1-3 under the induction of different methanol concentrations. VEGFA165-RBD was expressed in E. coli BL21 harboring pET28a( +)─VEGFA165-RBD vector under induction with IPTG with/without lactose. Interaction and biological activity of sKDR1-3 and VEGFA165-RBD were investigated by ELISA and anti-proliferation tests. sKDR1-3 migrated on SDS-PAGE gel as a 35-180 kDa protein due to glycosylation. The relative expression level of sKDR1-3 under 1% methanol was higher than 0.5% and 4% methanol induction. IPTG and cysteine were suitable for induction and refolding of VEGFA165-RBD. 25 ng sKDR1-3 and 20 ng VEGFA165-RBD showed strong binding. sKDR1-3 bound to VEGFA165-RBD and VEGF-A165 with dissociation constants of 0.148 and 0.2 nM, respectively. 4-10 nM concentrations of sKDR1-3 inhibited the proliferation of HUVE cells induced by 5 nM VEGFA165-RBD. In consideration, sKDR1-3 in the nanomolar concentration range, is a promising anticancer drug to inhibit angiogenesis.
Collapse
Affiliation(s)
- Zahra Fathi
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
- Department of Biology, Faculty of Sciences, University of Maragheh, Maragheh, Iran
| | | | - Reza H Sajedi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ehsan Dehnavi
- Gene Transfer Pioneers (GTP) Research Group, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohreh Jahanafrooz
- Department of Biology, Faculty of Sciences, University of Maragheh, Maragheh, Iran.
| |
Collapse
|
5
|
Brown NE, Ellerbe LR, Hollister SJ, Temenoff JS. Development and Characterization of Heparin-Containing Hydrogel/3D-Printed Scaffold Composites for Craniofacial Reconstruction. Ann Biomed Eng 2024; 52:2287-2307. [PMID: 38734845 DOI: 10.1007/s10439-024-03530-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
Regeneration of cartilage and bone tissues remains challenging in tissue engineering due to their complex structures, and the need for both mechanical support and delivery of biological repair stimuli. Therefore, the goal of this study was to develop a composite scaffold platform for anatomic chondral and osteochondral repair using heparin-based hydrogels to deliver small molecules within 3D-printed porous scaffolds that provide structure, stiffness, and controlled biologic delivery. We designed a mold-injection system to combine hydrolytically degradable hydrogels and 3D-printed scaffolds that could be employed rapidly (< 30 min) in operating room settings (~23 °C). Micro-CT analysis demonstrated the effectiveness of our injection system through homogeneously distributed hydrogel within the pores of the scaffolds. Hydrogels and composite scaffolds exhibited efficient loading (~94%) of a small positively charged heparin-binding molecule (crystal violet) with sustained release over 14 days and showed high viability of encapsulated porcine chondrocytes over 7 days. Compression testing demonstrated nonlinear viscoelastic behavior where tangent stiffness decreased with scaffold porosity (porous scaffold tangent stiffness: 70%: 4.9 MPa, 80%: 1.5 MPa, and 90%: 0.20 MPa) but relaxation was not affected. Lower-porosity scaffolds (70%) showed stiffness similar to lower ranges of trabecular bone (4-8 MPa) while higher-porosity scaffolds (80% and 90%) showed stiffness similar to auricular cartilage (0.16-2 MPa). Ultimately, this rapid composite scaffold fabrication method may be employed in the operating room and utilized to control biologic delivery within load-bearing scaffolds.
Collapse
Affiliation(s)
- Nettie E Brown
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr, Atlanta, GA, 30332, USA
| | - Lela R Ellerbe
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr, Atlanta, GA, 30332, USA
| | - Scott J Hollister
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr, Atlanta, GA, 30332, USA.
| | - Johnna S Temenoff
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr, Atlanta, GA, 30332, USA.
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr, Atlanta, GA, 30332, USA.
| |
Collapse
|
6
|
Song J, Shao L, Yu H, Meng C, Li G. Self-Assembly of Sulfate-Containing Peptides Sequesters VEGF for Inhibiting Cancer Cell Invasion. Biomacromolecules 2024; 25:3087-3097. [PMID: 38584438 DOI: 10.1021/acs.biomac.4c00168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Heparan sulfate proteoglycans (HSPGs) play a crucial role in regulating cancer growth and migration by mediating interactions with growth factors. In this study, we developed a self-assembling peptide (S1) containing a sulfate group to simulate the contiguous sulfated regions (S-domains) in heparan sulfate for growth factor binding, aiming to sequester growth factors like VEGF. Spectral and structural studies as well as simulation studies suggested that S1 self-assembled into nanostructures similar to the heparan sulfate chains and effectively bound to VEGF. On cancer cell surfaces, S1 self-assemblies sequestered VEGF, leading to a reduction in VEGF levels in the medium, consequently inhibiting cancer cell growth, invasion, and angiogenesis. This study highlights the potential of self-assembling peptides to emulate extracellular matrix functions, offering insights for future cancer therapeutic strategies.
Collapse
Affiliation(s)
- Jiaqi Song
- Department of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P. R. China
| | - Liang Shao
- Department of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P. R. China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Hongwen Yu
- Department of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P. R. China
| | - Caiting Meng
- Department of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P. R. China
| | - Guanying Li
- Department of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P. R. China
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P. R. China
| |
Collapse
|
7
|
Go YJ, Kalathingal M, Rhee YM. An Ensemble Docking Approach for Analyzing and Designing Aptamer Heterodimers Targeting VEGF 165. Int J Mol Sci 2024; 25:4066. [PMID: 38612876 PMCID: PMC11012306 DOI: 10.3390/ijms25074066] [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: 03/15/2024] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Vascular endothelial growth factor 165 (VEGF165) is a prominent isoform of the VEGF-A protein that plays a crucial role in various angiogenesis-related diseases. It is homodimeric, and each of its monomers is composed of two domains connected by a flexible linker. DNA aptamers, which have emerged as potent therapeutic molecules for many proteins with high specificity and affinity, can also work for VEGF165. A DNA aptamer heterodimer composed of monomers of V7t1 and del5-1 connected by a flexible linker (V7t1:del5-1) exhibits a greater binding affinity with VEGF165 compared to either of the two monomers alone. Although the structure of the complex formed between the aptamer heterodimer and VEGF165 is unknown due to the highly flexible linkers, gaining structural information will still be valuable for future developments. Toward this end of accessing structural information, we adopt an ensemble docking approach here. We first obtain an ensemble of structures for both VEGF165 and the aptamer heterodimer by considering both small- and large-scale motions. We then proceed through an extraction process based on ensemble docking, molecular dynamics simulations, and binding free energy calculations to predict the structures of the VEGF165/V7t1:del5-1 complex. Through the same procedures, we reach a new aptamer heterodimer that bears a locked nucleic acid-modified counterpart of V7t1, namely RNV66:del5-1, which also binds well with VEGF165. We apply the same protocol to the monomeric units V7t1, RNV66, and del5-1 to target VEGF165. We observe that V7t1:del5-1 and RNV66:del5-1 show higher binding affinities with VEGF165 than any of the monomers, consistent with experiments that support the notion that aptamer heterodimers are more effective anti-VEGF165 aptamers than monomeric aptamers. Among the five different aptamers studied here, the newly designed RNV66:del5-1 shows the highest binding affinity with VEGF165. We expect that our ensemble docking approach can help in de novo designs of homo/heterodimeric anti-angiogenic drugs to target the homodimeric VEGF165.
Collapse
Affiliation(s)
- Yeon Ju Go
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea;
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Mahroof Kalathingal
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea;
| | - Young Min Rhee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea;
| |
Collapse
|
8
|
Jian HJ, Anand A, Lai JY, Huang CC, Ma DHK, Lai CC, Chang HT. Ultrahigh-Efficacy VEGF Neutralization Using Carbonized Nanodonuts: Implications for Intraocular Anti-Angiogenic Therapy. Adv Healthc Mater 2024; 13:e2302881. [PMID: 38130100 DOI: 10.1002/adhm.202302881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/11/2023] [Indexed: 12/23/2023]
Abstract
Ocular angiogenesis, associated with diseases such as retinopathy of prematurity and diabetic retinopathy, is a leading cause of irreversible vision loss. Herein, carbon nanodonuts (CNDs) with a donut-shaped structure are synthesized using sodium alginate (SA) and 1,8-diaminooctane (DAO) through a one-step thermal process. The formation of SA/DAO-CNDs occurs through a crosslinking reaction between SA and DAO, creating amide bonds followed by partial carbonization. In human retinal pigment epithelial cells exposed to H2 O2 or lipopolysaccharide, the SA/DAO-CNDs display a more than fivefold reduction in reactive oxygen species and proinflammatory cytokines, such as IL-6 and IL-1β, when compared to carbonized nanomaterials produced exclusively from SA. Furthermore, the CNDs effectively inhibit vascular endothelial growth factor A-165 (VEGF-A165 )-induced cell migration and tube formation in human umbilical vein endothelial cells due to their strong affinity for VEGF-A165 , with a dissociation constant of 2.2 × 10-14 M, over 1600 times stronger than the commercial drug bevacizumab (Avastin). Trypsin digestion coupled with LC-MS/MS analysis reveals that VEGF-A165 interacts with SA/DAO-CNDs through its heparin-binding domain, leading to activity loss. The SA/DAO-CNDs demonstrate excellent biocompatibility and potent anti-angiogenic effects in chicken embryos and rabbit eyes. These findings suggest that SA/DAO-CNDs hold promise as a therapeutic agent for treating various angiogenesis-related ocular diseases.
Collapse
Affiliation(s)
- Hong-Jyuan Jian
- Department of Biomedical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Anisha Anand
- Department of Biomedical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Jui-Yang Lai
- Department of Biomedical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taoyuan, 33305, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, New Taipei City, 24301, Taiwan
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, 33303, Taiwan
| | - Chih-Ching Huang
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 20224, Taiwan
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung, 20224, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - David Hui-Kang Ma
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taoyuan, 33305, Taiwan
- Department of Chinese Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Chi-Chun Lai
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taoyuan, 33305, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Department of Ophthalmology, Chang Gung Memorial Hospital, Keelung, 20401, Taiwan
| | - Huan-Tsung Chang
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, 33302, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, 33302, Taiwan
- Center for Advanced Biomaterials and Technology Innovation, Chang Gung University, Taoyuan, 33302, Taiwan
- Division of Breast Surgery, Department of General Surgery, Chang Gung Memorial Hospital, Linkou, Taoyuan, 33305, Taiwan
| |
Collapse
|
9
|
Kalathingal M, Rhee YM. Molecular mechanism of binding between a therapeutic RNA aptamer and its protein target VEGF: A molecular dynamics study. J Comput Chem 2023; 44:1129-1137. [PMID: 36625560 DOI: 10.1002/jcc.27070] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 11/18/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023]
Abstract
Macugen is a therapeutic RNA aptamer against vascular endothelial growth factor (VEGF)-165, the VEGF isoform primarily responsible for angiogenesis. It has been reported that Macugen inhibits angiogenesis by specifically binding to the heparin binding domain (HBD) of VEGF165. The mechanism of the molecular recognition between HBD and Macugen is investigated here using all-atom molecular dynamics simulations. We find that Macugen recognizes HBD by an induced-fit mechanism with major conformational changes in Macugen and almost no changes in the structure of HBD, whereas HBD recognizes Macugen by a conformational selection mechanism.
Collapse
Affiliation(s)
- Mahroof Kalathingal
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Young Min Rhee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| |
Collapse
|
10
|
Heparin-Induced Changes of Vascular Endothelial Growth Factor (VEGF 165) Structure. Biomolecules 2023; 13:biom13010098. [PMID: 36671483 PMCID: PMC9856036 DOI: 10.3390/biom13010098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/28/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Vascular endothelial growth factor-A (VEGF-A), a secreted homodimeric glycoprotein, is a critical regulator of angiogenesis in normal and pathological states. The binding of heparin (HE) to VEGF165 (the major form of VEGF-A) modulates the angiogenesis-related cascade, but the mechanism of the observed changes at the structural level is still insufficiently explored. In the present study, we examined the effect of HE on the structural and physicochemical properties of recombinant human VEGF165 (rhVEGF165). The HE binding results in an increase of hydrophobic surface exposure in rhVEGF165 without changes in its secondary structure. Differential scanning calorimetry measurements for intact and HE-bound rhVEGF165 reveals the absence of any pronounced thermally induced transitions in the protein in the temperature range from 20 to 100 °C. The apolar area increase during the heparin binding explains the pronounced HE-induced oligomerization/aggregation of rhVEGF165, as studied by chemical glutaraldehyde cross-linking and dynamic light scattering. Molecular modeling and docking techniques were used to model the full structure of dimeric VEGF165 and to reveal putative molecular mechanisms underlying the function of the VEGF165/HE system. In general, the results obtained can be a basis for explaining the modulating effect of HE on the biological activity of VEGF-A.
Collapse
|
11
|
Spanou CES, Wohl AP, Doherr S, Correns A, Sonntag N, Lütke S, Mörgelin M, Imhof T, Gebauer JM, Baumann U, Grobe K, Koch M, Sengle G. Targeting of bone morphogenetic protein complexes to heparin/heparan sulfate glycosaminoglycans in bioactive conformation. FASEB J 2023; 37:e22717. [PMID: 36563024 DOI: 10.1096/fj.202200904r] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/25/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
Bone morphogenetic proteins (BMP) are powerful regulators of cellular processes such as proliferation, differentiation, and apoptosis. However, the specific molecular requirements controlling the bioavailability of BMPs in the extracellular matrix (ECM) are not yet fully understood. Our previous work showed that BMPs are targeted to the ECM as growth factor-prodomain (GF-PD) complexes (CPLXs) via specific interactions of their PDs. We showed that BMP-7 PD binding to the extracellular microfibril component fibrillin-1 renders the CPLXs from an open, bioactive V-shape into a closed, latent ring shape. Here, we show that specific PD interactions with heparin/heparan sulfate glycosaminoglycans (GAGs) allow to target and spatially concentrate BMP-7 and BMP-9 CPLXs in bioactive V-shape conformation. However, targeting to GAGs may be BMP specific, since BMP-10 GF and CPLX do not interact with heparin. Bioactivity assays on solid phase in combination with interaction studies showed that the BMP-7 PD protects the BMP-7 GF from inactivation by heparin. By using transmission electron microscopy, molecular docking, and site-directed mutagenesis, we determined the BMP-7 PD-binding site for heparin. Further, fine-mapping of the fibrillin-1-binding site within the BMP-7 PD and molecular modeling showed that both binding sites are mutually exclusive in the open V- versus closed ring-shape conformation. Together, our data suggest that targeting exquisite BMP PD-binding sites by extracellular protein and GAG scaffolds integrates BMP GF bioavailability in a contextual manner in development, postnatal life, and connective tissue disease.
Collapse
Affiliation(s)
- Chara E S Spanou
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Alexander P Wohl
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Sandra Doherr
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Annkatrin Correns
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Niklas Sonntag
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Steffen Lütke
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Matthias Mörgelin
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden.,Colzyx AB, Lund, Sweden
| | - Thomas Imhof
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Medical Faculty, Institute for Dental Research and Oral Musculoskeletal Biology, University of Cologne, Cologne, Germany
| | - Jan M Gebauer
- Institute of Biochemistry, University of Cologne, Cologne, Germany
| | - Ulrich Baumann
- Medical Faculty, Institute for Dental Research and Oral Musculoskeletal Biology, University of Cologne, Cologne, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Manuel Koch
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Medical Faculty, Institute for Dental Research and Oral Musculoskeletal Biology, University of Cologne, Cologne, Germany
| | - Gerhard Sengle
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), Cologne, Germany
| |
Collapse
|
12
|
Deering J, Lin DSY, D'Elia A, Zhang B, Grandfield K. Fabrication of succinate-alginate xerogel films for in vitro coupling of osteogenesis and neovascularization. BIOMATERIALS ADVANCES 2022; 141:213122. [PMID: 36162345 DOI: 10.1016/j.bioadv.2022.213122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/30/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
The osseointegration of metallic implants is reliant on a cascade of molecular interactions and the delivery of macromolecules to the implant environment that occurs before substantial bone formation. Early blood vessel formation is a requisite first step in the healing timeline for osteoid formation, where vascular development can be accelerated as a result of controlled hypoxic conditioning. In this study, alginate-derived xerogel films containing varied concentrations of disodium succinate salt which has been shown to induce pseudohypoxia (short-term hypoxic effects while maintaining an oxygenated environment) were developed. Xerogels were characterized for their morphology, succinate release over time and cellular response with osteoblast-mimicking Saos-2 and human umbilical vein endothelial cells (HUVEC). Scanning electron microscopy revealed a multiscale topography that may favour osseointegration and alamarBlue assays indicated no cytotoxic effects during in vitro proliferation of Saos-2 cells. pH measurements of eluted succinate reach 95 % of peak value after 7 h of immersion for all gels containing 10 mM of succinate or less, and 60 % within the first 40 min. In vitro exposure of HUVECs to succinate-conditioned media increased the net concentration of total proteins measured by bicinchoninic acid (BCA) assay and maintains stable vascular endothelial growth factor (VEGF) and extracellular platelet-derived growth factor (PDGF) for vessel formation through comparison of enzyme-linked immunosorbent assays (ELISAs) of the culture media and cell lysate. Tube formation assays also showed a sustained increase in tube diameter across the first 48 h of HUVEC culture when succinate concentrations of 1 and 10 μM in the xerogel. Overall, the succinate-alginate films serve as a prospective organic coating for bone-interfacing implant materials which may induce temporary pseudohypoxic conditions favourable for early angiogenesis and bone regeneration in vivo at succinate concentrations of 1 or 10 μM.
Collapse
Affiliation(s)
- Joseph Deering
- Department of Materials Science and Engineering, McMaster University, Hamilton, ON, Canada
| | - Dawn S Y Lin
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Andrew D'Elia
- Department of Materials Science and Engineering, McMaster University, Hamilton, ON, Canada
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada; School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
| | - Kathryn Grandfield
- Department of Materials Science and Engineering, McMaster University, Hamilton, ON, Canada; School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada; Brockhouse Institute for Materials Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
13
|
Inverting angiogenesis with interstitial flow and chemokine matrix-binding affinity. Sci Rep 2022; 12:4237. [PMID: 35273299 PMCID: PMC8913640 DOI: 10.1038/s41598-022-08186-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/15/2022] [Indexed: 11/30/2022] Open
Abstract
The molecular signaling pathways that orchestrate angiogenesis have been widely studied, but the role of biophysical cues has received less attention. Interstitial flow is unavoidable in vivo, and has been shown to dramatically change the neovascular patterns, but the mechanisms by which flow regulates angiogenesis remain poorly understood. Here, we study the complex interactions between interstitial flow and the affinity for matrix binding of different chemokine isoforms. Using a computational model, we find that changing the matrix affinity of the chemokine isoform can invert the effect of interstitial flow on angiogenesis—from preferential growth in the direction of the flow when the chemokine is initially matrix-bound to preferential flow against the flow when it is unbound. Although fluid forces signal endothelial cells directly, our data suggests a mechanism for the inversion based on biotransport arguments only, and offers a potential explanation for experimental results in which interstitial flow produced preferential vessel growth with and against the flow. Our results point to a particularly intricate effect of interstitial flow on angiogenesis in the tumor microenvironment, where the vessel network geometry and the interstitial flow patterns are complex.
Collapse
|
14
|
Ye X, Gaucher JF, Vidal M, Broussy S. A Structural Overview of Vascular Endothelial Growth Factors Pharmacological Ligands: From Macromolecules to Designed Peptidomimetics. Molecules 2021; 26:6759. [PMID: 34833851 PMCID: PMC8625919 DOI: 10.3390/molecules26226759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 12/27/2022] Open
Abstract
The vascular endothelial growth factor (VEGF) family of cytokines plays a key role in vasculogenesis, angiogenesis, and lymphangiogenesis. VEGF-A is the main member of this family, alongside placental growth factor (PlGF), VEGF-B/C/D in mammals, and VEGF-E/F in other organisms. To study the activities of these growth factors under physiological and pathological conditions, resulting in therapeutic applications in cancer and age-related macular degeneration, blocking ligands have been developed. These have mostly been large biomolecules like antibodies. Ligands with high affinities, at least in the nanomolar range, and accurate structural data from X-ray crystallography and NMR spectroscopy have been described. They constitute the main focus of this overview, which evidences similarities and differences in their binding modes. For VEGF-A ligands, and to a limited extent also for PlGF, a transition is now observed towards developing smaller ligands like nanobodies and peptides. These include unnatural amino acids and chemical modifications for designed and improved properties, such as serum stability and greater affinity. However, this review also highlights the scarcity of such small molecular entities and the striking lack of small organic molecule ligands. It also shows the gap between the rather large array of ligands targeting VEGF-A and the general absence of ligands binding other VEGF members, besides some antibodies. Future developments in these directions are expected in the upcoming years, and the study of these growth factors and their promising therapeutic applications will be welcomed.
Collapse
Affiliation(s)
- Xiaoqing Ye
- Faculté de Pharmacie de Paris, Université de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, 75006 Paris, France; (X.Y.); (M.V.)
| | - Jean-François Gaucher
- Laboratoire de Cristallographie et RMN Biologiques, Faculté de Pharmacie de Paris, Université de Paris, CiTCoM, 8038 CNRS, 75006 Paris, France;
| | - Michel Vidal
- Faculté de Pharmacie de Paris, Université de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, 75006 Paris, France; (X.Y.); (M.V.)
- Service Biologie du Médicament, Toxicologie, AP-HP, Hôpital Cochin, 75014 Paris, France
| | - Sylvain Broussy
- Faculté de Pharmacie de Paris, Université de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, 75006 Paris, France; (X.Y.); (M.V.)
| |
Collapse
|
15
|
The Chemokine-Based Peptide, CXCL9(74-103), Inhibits Angiogenesis by Blocking Heparan Sulfate Proteoglycan-Mediated Signaling of Multiple Endothelial Growth Factors. Cancers (Basel) 2021; 13:cancers13205090. [PMID: 34680238 PMCID: PMC8534003 DOI: 10.3390/cancers13205090] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Major angiogenic growth factors activate downstream signaling cascades by interacting with both receptor tyrosine kinases (RTKs) and cell surface proteoglycans, such as heparan sulfate proteoglycans (HSPGs). As current anti-angiogenesis regimens in cancer are often faced with resistance, alternative therapeutic strategies are highly needed. The aim of our study was to investigate the impact on angiogenic signaling when we interfered with growth factor-HSPG interactions using a CXCL9 chemokine-derived peptide with high affinity for HS. Abstract Growth factors such as vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF) and epidermal growth factor (EGF) are important angiogenesis-mediating factors. They exert their effects not only through their respective receptor tyrosine kinases (RTKs), but they also require molecular pairing with heparan sulfate proteoglycans (HSPGs). Angiogenic growth factors and their signaling pathways are commonly targeted in current anti-angiogenic cancer therapies but have unfortunately insufficient impact on patient survival. Considering their obvious role in pathological angiogenesis, HS-targeting drugs have become an appealing new strategy. Therefore, we aimed to reduce angiogenesis through interference with growth factor-HS binding and downstream signaling using a CXCL9-derived peptide with a high affinity for glycosaminoglycans (GAGs), CXCL9(74-103). We showed that CXCL9(74-103) reduced EGF-, VEGF165- and FGF-2-mediated angiogenic processes in vitro, such as endothelial cell proliferation, chemotaxis, adhesion and sprouting, without exerting cell toxicity. CXCL9(74-103) interfered with growth factor signaling in diverse ways, e.g., by diminishing VEGF165 binding to HS and by direct association with FGF-2. The dependency of CXCL9(74-103) on HS for binding to HMVECs and for exerting its anti-angiogenic activity was also demonstrated. In vivo, CXCL9(74-103) attenuated neovascularization in the Matrigel plug assay, the corneal cauterization assay and in MDA-MB-231 breast cancer xenografts. Additionally, CXCL9(74-103) reduced vascular leakage in the retina of diabetic rats. In contrast, CXCL9(86-103), a peptide with low GAG affinity, showed no overall anti-angiogenic activity. Altogether, our results indicate that CXCL9(74-103) reduces angiogenesis by interfering with multiple HS-dependent growth factor signaling pathways.
Collapse
|
16
|
Banik N, Yang SB, Kang TB, Lim JH, Park J. Heparin and Its Derivatives: Challenges and Advances in Therapeutic Biomolecules. Int J Mol Sci 2021; 22:ijms221910524. [PMID: 34638867 PMCID: PMC8509054 DOI: 10.3390/ijms221910524] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Heparin has been extensively studied as a safe medicine and biomolecule over the past few decades. Heparin derivatives, including low-molecular-weight heparins (LMWH) and heparin pentasaccharide, are effective anticoagulants currently used in clinical settings. They have also been studied as functional biomolecules or biomaterials for various therapeutic uses to treat diseases. Heparin, which has a similar molecular structure to heparan sulfate, can be used as a remarkable biomedicine due to its uniquely high safety and biocompatibility. In particular, it has recently drawn attention for use in drug-delivery systems, biomaterial-based tissue engineering, nanoformulations, and new drug-development systems through molecular formulas. A variety of new heparin-based biomolecules and conjugates have been developed in recent years and are currently being evaluated for use in clinical applications. This article reviews heparin derivatives recently studied in the field of drug development for the treatment of various diseases.
Collapse
Affiliation(s)
- Nipa Banik
- Department of Integrated Biosciences, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (N.B.); (S.-B.Y.); (T.-B.K.); (J.-H.L.)
| | - Seong-Bin Yang
- Department of Integrated Biosciences, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (N.B.); (S.-B.Y.); (T.-B.K.); (J.-H.L.)
| | - Tae-Bong Kang
- Department of Integrated Biosciences, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (N.B.); (S.-B.Y.); (T.-B.K.); (J.-H.L.)
| | - Ji-Hong Lim
- Department of Integrated Biosciences, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (N.B.); (S.-B.Y.); (T.-B.K.); (J.-H.L.)
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Korea
| | - Jooho Park
- Department of Integrated Biosciences, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (N.B.); (S.-B.Y.); (T.-B.K.); (J.-H.L.)
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Korea
- Correspondence:
| |
Collapse
|
17
|
Peptide Inhibitors of Vascular Endothelial Growth Factor A: Current Situation and Perspectives. Pharmaceutics 2021; 13:pharmaceutics13091337. [PMID: 34575413 PMCID: PMC8467741 DOI: 10.3390/pharmaceutics13091337] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Vascular endothelial growth factors (VEGFs) are the family of extracellular signaling proteins involved in the processes of angiogenesis. VEGFA overexpression and altered regulation of VEGFA signaling pathways lead to pathological angiogenesis, which contributes to the progression of various diseases, such as age-related macular degeneration and cancer. Monoclonal antibodies and decoy receptors have been extensively used in the anti-angiogenic therapies for the neutralization of VEGFA. However, multiple side effects, solubility and aggregation issues, and the involvement of compensatory VEGFA-independent pro-angiogenic mechanisms limit the use of the existing VEGFA inhibitors. Short chemically synthesized VEGFA binding peptides are a promising alternative to these full-length proteins. In this review, we summarize anti-VEGFA peptides identified so far and discuss the molecular basis of their inhibitory activity to highlight their pharmacological potential as anti-angiogenic drugs.
Collapse
|
18
|
Bornert F, Clauss F, Hua G, Idoux-Gillet Y, Keller L, Fernandez De Grado G, Offner D, Smaida R, Wagner Q, Fioretti F, Kuchler-Bopp S, Schulz G, Wenzel W, Gentile L, Risser L, Müller B, Huck O, Benkirane-Jessel N. Mechanistic Illustration: How Newly-Formed Blood Vessels Stopped by the Mineral Blocks of Bone Substitutes Can Be Avoided by Using Innovative Combined Therapeutics. Biomedicines 2021; 9:952. [PMID: 34440156 PMCID: PMC8394928 DOI: 10.3390/biomedicines9080952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/16/2021] [Accepted: 08/01/2021] [Indexed: 12/30/2022] Open
Abstract
One major limitation for the vascularization of bone substitutes used for filling is the presence of mineral blocks. The newly-formed blood vessels are stopped or have to circumvent the mineral blocks, resulting in inefficient delivery of oxygen and nutrients to the implant. This leads to necrosis within the implant and to poor engraftment of the bone substitute. The aim of the present study is to provide a bone substitute currently used in the clinic with suitably guided vascularization properties. This therapeutic hybrid bone filling, containing a mineral and a polymeric component, is fortified with pro-angiogenic smart nano-therapeutics that allow the release of angiogenic molecules. Our data showed that the improved vasculature within the implant promoted new bone formation and that the newly-formed bone swapped the mineral blocks of the bone substitutes much more efficiently than in non-functionalized bone substitutes. Therefore, we demonstrated that our therapeutic bone substitute is an advanced therapeutical medicinal product, with great potential to recuperate and guide vascularization that is stopped by mineral blocks, and can improve the regeneration of critical-sized bone defects. We have also elucidated the mechanism to understand how the newly-formed vessels can no longer encounter mineral blocks and pursue their course of vasculature, giving our advanced therapeutical bone filling great potential to be used in many applications, by combining filling and nano-regenerative medicine that currently fall short because of problems related to the lack of oxygen and nutrients.
Collapse
Affiliation(s)
- Fabien Bornert
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - François Clauss
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Guoqiang Hua
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Ysia Idoux-Gillet
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Laetitia Keller
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Gabriel Fernandez De Grado
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Damien Offner
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Rana Smaida
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Quentin Wagner
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Florence Fioretti
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Sabine Kuchler-Bopp
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Georg Schulz
- Biomaterials Science Center, University of Basel, Gewerbestrasse 14, CH-4123 Allschwil, Switzerland; (G.S.); (B.M.)
| | - Wolfgang Wenzel
- Institute of Nanotechnology, Karlsruhe Institute of Technology, Campus North, Building 640, DE-76131 Karlsruhe, Germany;
| | - Luca Gentile
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Laurent Risser
- Toulouse Institute of Mathematics, UMR 5219 University of Toulouse, CNRS UPS IMT, 31062 Toulouse, France;
| | - Bert Müller
- Biomaterials Science Center, University of Basel, Gewerbestrasse 14, CH-4123 Allschwil, Switzerland; (G.S.); (B.M.)
| | - Olivier Huck
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| |
Collapse
|
19
|
Zhang J, Liu X, Ma K, Chen M, Xu H, Niu X, Gu H, Wang R, Chen X, Sun H. Collagen/heparin scaffold combined with vascular endothelial growth factor promotes the repair of neurological function in rats with traumatic brain injury. Biomater Sci 2021; 9:745-764. [DOI: 10.1039/c9bm01446b] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The objective of this study was to evaluate the therapy effects of a novel biological scaffold containing heparin, collagen and vascular endothelial growth factor (VEGF) in treating traumatic brain injury (TBI).
Collapse
Affiliation(s)
- Jian Zhang
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| | - Xiaoyin Liu
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| | - Ke Ma
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| | - Miao Chen
- Affiliated Hospital of Traditional Chinese Medicine
- Xinjiang Medical University
- Urumqi
- China
| | - Huiyou Xu
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| | | | - Haoran Gu
- The 947th hospital of Chinese People's Liberation Army
- Xinjiang
- China
| | - Renjie Wang
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| | - Xuyi Chen
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| | - HongTao Sun
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| |
Collapse
|
20
|
Lee W, Park JW, Go YJ, Kim WJ, Rhee YM. Considering both small and large scale motions of vascular endothelial growth factor (VEGF) is crucial for reliably predicting its binding affinities to DNA aptamers. RSC Adv 2021; 11:9315-9326. [PMID: 35423456 PMCID: PMC8695334 DOI: 10.1039/d0ra10106k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/23/2021] [Indexed: 11/21/2022] Open
Abstract
Considering both small and large scale motions of VEGF is crucial to predict its relative binding affinities to DNA aptamer variants with docking.
Collapse
Affiliation(s)
- Wook Lee
- Department of Chemistry
- Pohang University of Science and Technology (POSTECH)
- Pohang 37673
- Korea
- Department of Chemistry
| | - Jae Whee Park
- Department of Chemistry
- Korea Advanced Institute of Science and Technology (KAIST)
- Daejeon 34141
- Korea
| | - Yeon Ju Go
- Department of Chemistry
- Korea Advanced Institute of Science and Technology (KAIST)
- Daejeon 34141
- Korea
| | - Won Jong Kim
- Department of Chemistry
- Pohang University of Science and Technology (POSTECH)
- Pohang 37673
- Korea
| | - Young Min Rhee
- Department of Chemistry
- Korea Advanced Institute of Science and Technology (KAIST)
- Daejeon 34141
- Korea
| |
Collapse
|
21
|
Molecular Targeting of VEGF with a Suramin Fragment-DOCA Conjugate by Mimicking the Action of Low Molecular Weight Heparins. Biomolecules 2020; 11:biom11010046. [PMID: 33396366 PMCID: PMC7823656 DOI: 10.3390/biom11010046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 01/01/2023] Open
Abstract
Molecular targeting of growth factors has shown great therapeutic potential in pharmaceutical research due to their roles in pathological conditions. In the present study, we developed a novel suramin fragment and deoxycholic acid conjugate (SFD) that exhibited the potential to bind to the heparin-binding site (HBD) of vascular endothelial growth factor (VEGF) and to inhibit its pathogenic action for the first time. Notably, SFD was optimally designed for binding to the HBD of VEGF using the naphthalenetrisulfonate group, allowing to observe its excellent binding efficacy in a surface plasmon resonance (SPR) study, showing remarkable binding affinity (KD = 3.8 nM) as a small molecule inhibitor. In the tubular formation assay, it was observed that SFD could bind to HBD and exhibit antiangiogenic efficacy by inhibiting VEGF, such as heparins. The cellular treatment of SFD resulted in VEGF-inhibitory effects in human umbilical vein endothelial cells (HUVECs). Therefore, we propose that SFD can be employed as a novel drug candidate to inhibit the pathophysiological action of VEGF in diseases. Consequently, SFD, which has a molecular structure optimized for binding to HBD, is put forward as a new chemical VEGF inhibitor.
Collapse
|
22
|
Puszko AK, Sosnowski P, Rignault-Bricard R, Hermine O, Hopfgartner G, Pułka-Ziach K, Lepelletier Y, Misicka A. Urea-Peptide Hybrids as VEGF-A 165/NRP-1 Complex Inhibitors with Improved Receptor Affinity and Biological Properties. Int J Mol Sci 2020; 22:ijms22010072. [PMID: 33374715 PMCID: PMC7793531 DOI: 10.3390/ijms22010072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 12/31/2022] Open
Abstract
Neuropilin-1 (NRP-1), the major co-receptor of vascular endothelial growth factor receptor-2 (VEGFR-2), may also independently act with VEGF-A165 to stimulate tumour growth and metastasis. Therefore, there is great interest in compounds that can block VEGF-A165/NRP-1 interaction. Peptidomimetic type inhibitors represent a promising strategy in the treatment of NRP-1-related disorders. Here, we present the synthesis, affinity, enzymatic stability, molecular modeling and in vitro binding evaluation of the branched urea–peptide hybrids, based on our previously reported Lys(hArg)-Dab-Oic-Arg active sequence, where the Lys(hArg) branching has been modified by introducing urea units to replace the peptide bond at various positions. One of the resulting hybrids increased the affinity of the compound for NRP-1 more than 10-fold, while simultaneously improving resistance for proteolytic stability in serum. In addition, ligand binding to NRP-1 induced rapid protein stock exocytotic trafficking to the plasma membrane in breast cancer cells. Examined properties characterize this compound as a good candidate for further development of VEGF165/NRP-1 inhibitors.
Collapse
Affiliation(s)
- Anna K. Puszko
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland;
- Correspondence: (A.K.P.); (A.M.)
| | - Piotr Sosnowski
- Department of Inorganic and Analytical Chemistry, University of Geneva, 24 Quai Ernest Ansermet, CH-1211 Geneva, Switzerland; (P.S.); (G.H.)
| | - Rachel Rignault-Bricard
- Imagine Institute, Université de Paris, 24 boulevard Montparnasse, 75015 Paris, France; (R.R.-B.); (O.H.); (Y.L.)
- INSERM UMR 1163, Laboratory of Cellular and Molecular Basis of Normal Hematopoiesis and Hematological Disorders: Therapeutical Implications, 24 Boulevard Montparnasse, 75015 Paris, France
| | - Olivier Hermine
- Imagine Institute, Université de Paris, 24 boulevard Montparnasse, 75015 Paris, France; (R.R.-B.); (O.H.); (Y.L.)
- INSERM UMR 1163, Laboratory of Cellular and Molecular Basis of Normal Hematopoiesis and Hematological Disorders: Therapeutical Implications, 24 Boulevard Montparnasse, 75015 Paris, France
| | - Gérard Hopfgartner
- Department of Inorganic and Analytical Chemistry, University of Geneva, 24 Quai Ernest Ansermet, CH-1211 Geneva, Switzerland; (P.S.); (G.H.)
| | | | - Yves Lepelletier
- Imagine Institute, Université de Paris, 24 boulevard Montparnasse, 75015 Paris, France; (R.R.-B.); (O.H.); (Y.L.)
- INSERM UMR 1163, Laboratory of Cellular and Molecular Basis of Normal Hematopoiesis and Hematological Disorders: Therapeutical Implications, 24 Boulevard Montparnasse, 75015 Paris, France
| | - Aleksandra Misicka
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland;
- Department of Neuropeptides, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland
- Correspondence: (A.K.P.); (A.M.)
| |
Collapse
|
23
|
Manochehry S, Gu J, McConnell EM, Salena BJ, Li Y. In Vitro Selection of New DNA Aptamers for Human Vascular Endothelial Growth Factor 165. Chembiochem 2020; 21:2029-2036. [PMID: 32180322 DOI: 10.1002/cbic.202000024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/26/2020] [Indexed: 11/06/2022]
Abstract
Two DNA aptamers that bind the heparin-binding domain (HBD) of the human vascular endothelial growth factor 165 (VEGF-165) have been previously reported. Although VEGF-165 is a homodimeric protein and the two aptamers have different sequences and secondary structures, the aptamers appear to occupy the same binding site and cannot form a 2 : 1 aptamer/protein complex, thus making them unsuitable for creating a higher-affinity dimeric DNA aptamer. This has motivated us to conduct a new in vitro selection experiment to search for new VEGF-165-binding DNA aptamers with different properties. We undertook a multistream selection strategy in which the concentration of VEGF-165 was varied significantly. We carried out 11 rounds of selection, and next-generation sequencing was conducted for every round in each stream. From comprehensive sequence analysis, we identified four classes of DNA aptamers, of which two were reported before, but two are new DNA aptamers. One of the new aptamers exhibits a unique property that has never been observed before: it is capable of forming the 2 : 1 aptamer/protein complex with VEGF-165. This work has expanded the repertoire of VEGF-165-binding DNA aptamers and creates a possibility to engineer a higher affinity homodimeric aptamer for VEGF-165.
Collapse
Affiliation(s)
- Sepehr Manochehry
- M.G. DeGroote Institute for Infectious Disease Research Department of Biochemistry and Biomedical Sciences DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4 K1, Canada
| | - Jimmy Gu
- M.G. DeGroote Institute for Infectious Disease Research Department of Biochemistry and Biomedical Sciences DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4 K1, Canada
| | - Erin M McConnell
- M.G. DeGroote Institute for Infectious Disease Research Department of Biochemistry and Biomedical Sciences DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4 K1, Canada
| | - Bruno J Salena
- Department of Medicine DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4 K1, Canada
| | - Yingfu Li
- M.G. DeGroote Institute for Infectious Disease Research Department of Biochemistry and Biomedical Sciences DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4 K1, Canada
| |
Collapse
|
24
|
Puszko AK, Sosnowski P, Raynaud F, Hermine O, Hopfgartner G, Lepelletier Y, Misicka A. Does Cysteine Rule (CysR) Complete the CendR Principle? Increase in Affinity of Peptide Ligands for NRP-1 Through the Presence of N-Terminal Cysteine. Biomolecules 2020; 10:biom10030448. [PMID: 32183142 PMCID: PMC7175122 DOI: 10.3390/biom10030448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/23/2020] [Accepted: 03/05/2020] [Indexed: 01/13/2023] Open
Abstract
The structure-activity relationship of branched H-Lys(hArg)-Dab-Dhp-Arg-OH sequence analogues, modified with Cys-Asp or Cys at N-terminal amino acids (Lys, hArg), in VEGF-A165/Neuropilin-1 complex inhibition is presented. The addition of Cys residue led to a 100-fold decrease in the IC50 value, compared to the parent peptide. The change occurred regardless of coupling Cys to the free N-terminal amino group present in the main or the side chain. A few analogues extended by the attachment of Cys at the N-terminus of several potent NRP-1 peptide ligands documented in the literature are also presented. In all studied cases, the enhancement of inhibitory properties after the addition of Cys at the N-terminus is observed. It is particularly evident for the tetrapeptide derived from the C-terminus of VEGF-A165 (KPRR), suggesting that extending the K/RXXK/R motif (CendR) with the Cys moiety can significantly improve affinity to NRP-1 of CendR peptides.
Collapse
Affiliation(s)
- Anna K. Puszko
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland;
| | - Piotr Sosnowski
- Department of Inorganic and Analytical Chemistry, University of Geneva, 24 Quai Ernest Ansermet, CH-1211 4 Geneva, Switzerland; (P.S.); (G.H.)
| | - Françoise Raynaud
- Imagine Institute, Université de Paris, 24 boulevard Montparnasse, 75015 Paris, France; (F.R.); (O.H.)
- INSERM UMR 1163, Laboratory of Cellular and Molecular Basis of Normal Hematopoiesis and Hematological Disorders: Therapeutical Implications, 24 boulevard Montparnasse, 75015 Paris, France
- CNRS ERL 8254, 24 boulevard Montparnasse, 75015 Paris, France
| | - Olivier Hermine
- Imagine Institute, Université de Paris, 24 boulevard Montparnasse, 75015 Paris, France; (F.R.); (O.H.)
- INSERM UMR 1163, Laboratory of Cellular and Molecular Basis of Normal Hematopoiesis and Hematological Disorders: Therapeutical Implications, 24 boulevard Montparnasse, 75015 Paris, France
- CNRS ERL 8254, 24 boulevard Montparnasse, 75015 Paris, France
| | - Gérard Hopfgartner
- Department of Inorganic and Analytical Chemistry, University of Geneva, 24 Quai Ernest Ansermet, CH-1211 4 Geneva, Switzerland; (P.S.); (G.H.)
| | - Yves Lepelletier
- Imagine Institute, Université de Paris, 24 boulevard Montparnasse, 75015 Paris, France; (F.R.); (O.H.)
- INSERM UMR 1163, Laboratory of Cellular and Molecular Basis of Normal Hematopoiesis and Hematological Disorders: Therapeutical Implications, 24 boulevard Montparnasse, 75015 Paris, France
- CNRS ERL 8254, 24 boulevard Montparnasse, 75015 Paris, France
- Correspondence: (Y.L.); (A.M.); Tel.: +33-14275-4283 (Y.L.); +48-22-552-6424 (A.M.)
| | - Aleksandra Misicka
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland;
- Department of Neuropeptides, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland
- Correspondence: (Y.L.); (A.M.); Tel.: +33-14275-4283 (Y.L.); +48-22-552-6424 (A.M.)
| |
Collapse
|
25
|
Dual Action of Sulfated Hyaluronan on Angiogenic Processes in Relation to Vascular Endothelial Growth Factor-A. Sci Rep 2019; 9:18143. [PMID: 31792253 PMCID: PMC6889296 DOI: 10.1038/s41598-019-54211-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/05/2019] [Indexed: 01/13/2023] Open
Abstract
Pathological healing characterized by abnormal angiogenesis presents a serious burden to patients’ quality of life requiring innovative treatment strategies. Glycosaminoglycans (GAG) are important regulators of angiogenic processes. This experimental and computational study revealed how sulfated GAG derivatives (sGAG) influence the interplay of vascular endothelial growth factor (VEGF)165 and its heparin-binding domain (HBD) with the signaling receptor VEGFR-2 up to atomic detail. There was profound evidence for a HBD-GAG-HBD stacking configuration. Here, the sGAG act as a “molecular glue” leading to recognition modes in which sGAG interact with two VEGF165-HBDs. A 3D angiogenesis model demonstrated the dual regulatory role of high-sulfated derivatives on the biological activity of endothelial cells. While GAG alone promote sprouting, they downregulate VEGF165-mediated signaling and, thereby, elicit VEGF165-independent and -dependent effects. These findings provide novel insights into the modulatory potential of sGAG derivatives on angiogenic processes and point towards their prospective application in treating abnormal angiogenesis.
Collapse
|
26
|
Manochehry S, McConnell EM, Li Y. Unraveling Determinants of Affinity Enhancement in Dimeric Aptamers for a Dimeric Protein. Sci Rep 2019; 9:17824. [PMID: 31780794 PMCID: PMC6883073 DOI: 10.1038/s41598-019-54005-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 09/20/2019] [Indexed: 11/09/2022] Open
Abstract
High-affinity aptamers can be derived de novo by using stringent conditions in SELEX (Systematic Evolution of Ligands by EXponential enrichment) experiments or can be engineered post SELEX via dimerization of selected aptamers. Using electrophoretic mobility shift assays, we studied a series of heterodimeric and homodimeric aptamers, constructed from two DNA aptamers with distinct primary sequences and secondary structures, previously isolated for VEGF-165, a homodimeric protein. We investigated four factors envisaged to impact the affinity of a dimeric aptamer to a dimeric protein: (1) length of the linker between two aptamer domains, (2) linking orientation, (3) binding-site compatibility of two component aptamers in a heterodimeric aptamer, and (4) steric acceptability of the two identical aptamers in a homodimeric aptamer. All heterodimeric aptamers for VEGF-165 were found to exhibit monomeric aptamer-like affinity and the lack of affinity enhancement was attributed to binding-site overlap by the constituent aptamers. The best homodimeric aptamer showed 2.8-fold better affinity than its monomeric unit (Kd = 13.6 ± 2.7 nM compared to 37.9 ± 14 nM), however the barrier to further affinity enhancement was ascribed to steric interference of the constituent aptamers. Our findings point to the need to consider the issues of binding-site compatibility and spatial requirement of aptamers for the development of dimeric aptamers capable of bivalent recognition. Thus, determinants highlighted herein should be assessed in future multimerization efforts.
Collapse
Affiliation(s)
- Sepehr Manochehry
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St. W., Hamilton, ON, L8S 4K1, Canada
| | - Erin M McConnell
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St. W., Hamilton, ON, L8S 4K1, Canada
| | - Yingfu Li
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St. W., Hamilton, ON, L8S 4K1, Canada. .,Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St. W., Hamilton, ON, L8S 4K1, Canada.
| |
Collapse
|
27
|
Wei SC, Chang L, Huang CC, Chang HT. Dual-functional gold nanoparticles with antimicrobial and proangiogenic activities improve the healing of multidrug-resistant bacteria-infected wounds in diabetic mice. Biomater Sci 2019; 7:4482-4490. [PMID: 31531425 DOI: 10.1039/c9bm00772e] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Gold nanoparticles (Au NPs) are conjugated with the vascular endothelial growth factor-A165 (VEGF-A165) and (11-mercaptoundecyl)-N,N,N-trimethylammonium (11-MTA) cation to form dual-functional gold nanoparticles (11-MTA/VEGF-Au NPs) that possess antimicrobial and proangiogenic activities for wound healing in diabetic (db/db) mice. VEGF-A165 is a popular proangiogenic growth factor that stimulates multiple components in the wound-healing cascade. On the other hand, 11-MTA possesses antibacterial activity and can be bound to Au NPs easily through Au-S bonding. We have found that the surface density of VEGF-A165 plays a vital role in promoting the proliferation, migration, and tube formation of human umbilical vein endothelial cells. 11-MTA tethered on the VEGF-modified Au NPs enables the nanocomposites (i.e., 11-MTA/VEGF-Au NPs) to exhibit a strong antimicrobial activity against multidrug-resistant bacteria [methicillin-resistant S. aureus (MRSA)]. The minimal inhibition concentration of 11-MTA/VEGF-Au NPs is ∼450-fold lower than that of 11-MTA, revealing their high antibacterial efficiency. 11-MTA/VEGF-Au NPs exhibit high biocompatibility. 11-MTA/VEGF-Au NPs as dressing materials to treat MRSA-infected wounds in diabetic mice not only show strong in vivo bactericidal activities but also enhance the healing process of the formation of collagen fibers and epithelialization. Our results show that dual-functional 11-MTA/VEGF-Au NPs are promising agents for clinical applications like treating chronic wound infections.
Collapse
Affiliation(s)
- Shih-Chun Wei
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.
| | - Lung Chang
- Department of Pediatrics, Mackay Memorial Hospital and Mackay Junior College of Medicine, Nursing and Management, Taipei, 10449, Taiwan
| | - Chih-Ching Huang
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 20224, Taiwan. and Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung, 20224, Taiwan and School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Huan-Tsung Chang
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan. and Department of Chemistry, Chung Yuan Christian University, Taoyuan City 32023, Taiwan
| |
Collapse
|
28
|
Puszko AK, Sosnowski P, Pułka-Ziach K, Hermine O, Hopfgartner G, Lepelletier Y, Misicka A. Urea moiety as amide bond mimetic in peptide-like inhibitors of VEGF-A 165/NRP-1 complex. Bioorg Med Chem Lett 2019; 29:2493-2497. [PMID: 31326342 DOI: 10.1016/j.bmcl.2019.07.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/07/2019] [Accepted: 07/08/2019] [Indexed: 12/23/2022]
Abstract
NRP-1 is an important co-receptor of vascular endothelial growth factor receptor-2 (VEGFR-2). Many reports suggested that NRP-1 might also serve as a separate receptor for VEGF-A165 causing stimulation of tumour growth and metastasis. Therefore, compounds interfering with VEGF-A165/NRP-1 complex triggered interest in the design of new molecules, including peptides, as anti-angiogenic and anti-tumour drugs. Here, we report the synthesis, affinity and stability evaluation of the urea-peptide hybrids, based on general Lys(hArg)-AA2-AA3-Arg sequence, where hArg residue was substituted by Arg urea unit. Such substitution does not substantially affected affinity of compounds for NRP-1 but significantly increased their proteolytic stability in plasma.
Collapse
Affiliation(s)
- Anna K Puszko
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland.
| | - Piotr Sosnowski
- Department of Inorganic and Analytical Chemistry, University of Geneva, 24 Quai Ernest Ansermet, CH-1211 Geneva 4, Switzerland; Department of Neuropeptides, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland
| | | | - Olivier Hermine
- Université de Paris, Imagine Institute, 24 Boulevard Montparnasse, 75015 Paris, France; INSERM UMR 1163, Laboratory of Cellular and Molecular Basis of Normal Hematopoiesis and Hematological Disorders: Therapeutical Implications, 24 Boulevard Montparnasse, 75015 Paris, France; CNRS ERL 8254, 24 Boulevard Montparnasse, 75015 Paris, France
| | - Gérard Hopfgartner
- Department of Inorganic and Analytical Chemistry, University of Geneva, 24 Quai Ernest Ansermet, CH-1211 Geneva 4, Switzerland
| | - Yves Lepelletier
- Université de Paris, Imagine Institute, 24 Boulevard Montparnasse, 75015 Paris, France; INSERM UMR 1163, Laboratory of Cellular and Molecular Basis of Normal Hematopoiesis and Hematological Disorders: Therapeutical Implications, 24 Boulevard Montparnasse, 75015 Paris, France; CNRS ERL 8254, 24 Boulevard Montparnasse, 75015 Paris, France
| | - Aleksandra Misicka
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland; Department of Neuropeptides, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland.
| |
Collapse
|
29
|
Engineered delivery strategies for enhanced control of growth factor activities in wound healing. Adv Drug Deliv Rev 2019; 146:190-208. [PMID: 29879493 DOI: 10.1016/j.addr.2018.06.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/18/2018] [Accepted: 06/01/2018] [Indexed: 12/18/2022]
Abstract
Growth factors (GFs) are versatile signalling molecules that orchestrate the dynamic, multi-stage process of wound healing. Delivery of exogenous GFs to the wound milieu to mediate healing in an active, physiologically-relevant manner has shown great promise in laboratories; however, the inherent instability of GFs, accompanied with numerous safety, efficacy and cost concerns, has hindered the clinical success of GF delivery. In this article, we highlight that the key to overcoming these challenges is to enhance the control of the activities of GFs throughout the delivering process. We summarise the recent strategies based on biomaterials matrices and molecular engineering, which aim to improve the conditions of GFs for delivery (at the 'supply' end of the delivery), increase the stability and functions of GFs in extracellular matrix (in transportation to target cells), as well as enhance the GFs/receptor interaction on the cell membrane (at the 'destination' end of the delivery). Many of these investigations have led to encouraging outcomes in various in vitro and in vivo regenerative models with considerable translational potential.
Collapse
|
30
|
Leuning DG, Witjas FMR, Maanaoui M, de Graaf AMA, Lievers E, Geuens T, Avramut CM, Wiersma LE, van den Berg CW, Sol WMPJ, de Boer H, Wang G, LaPointe VLS, van der Vlag J, van Kooten C, van den Berg BM, Little MH, Engelse MA, Rabelink TJ. Vascular bioengineering of scaffolds derived from human discarded transplant kidneys using human pluripotent stem cell-derived endothelium. Am J Transplant 2019; 19:1328-1343. [PMID: 30506641 PMCID: PMC6590331 DOI: 10.1111/ajt.15200] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 10/22/2018] [Accepted: 11/21/2018] [Indexed: 01/25/2023]
Abstract
The bioengineering of a replacement kidney has been proposed as an approach to address the growing shortage of donor kidneys for the treatment of chronic kidney disease. One approach being investigated is the recellularization of kidney scaffolds. In this study, we present several key advances toward successful re-endothelialization of whole kidney matrix scaffolds from both rodents and humans. Based on the presence of preserved glycosoaminoglycans within the decelullarized kidney scaffold, we show improved localization of delivered endothelial cells after preloading of the vascular matrix with vascular endothelial growth factor and angiopoietin 1. Using a novel simultaneous arteriovenous delivery system, we report the complete re-endothelialization of the kidney vasculature, including the glomerular and peritubular capillaries, using human inducible pluripotent stem cell -derived endothelial cells. Using this source of endothelial cells, it was possible to generate sufficient endothelial cells to recellularize an entire human kidney scaffold, achieving efficient cell delivery, adherence, and endothelial cell proliferation and survival. Moreover, human re-endothelialized scaffold could, in contrast to the non-re-endothelialized human scaffold, be fully perfused with whole blood. These major advances move the field closer to a human bioengineered kidney.
Collapse
Affiliation(s)
- Daniëlle G. Leuning
- Department of NephrologyLeiden University Medical CenterLeidenThe Netherlands
| | - Franca M. R. Witjas
- Department of NephrologyLeiden University Medical CenterLeidenThe Netherlands
| | - Mehdi Maanaoui
- Department of NephrologyLeiden University Medical CenterLeidenThe Netherlands,Nephrology DepartmentUniversity of LilleCHU LilleF‐59000LilleFrance
| | | | - Ellen Lievers
- Department of NephrologyLeiden University Medical CenterLeidenThe Netherlands
| | - Thomas Geuens
- MERLN Institute for Technology–Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Christina M. Avramut
- Department of Molecular Cell BiologySection Electron MicroscopyLeiden University Medical CenterLeidenThe Netherlands
| | - Loes E. Wiersma
- Department of NephrologyLeiden University Medical CenterLeidenThe Netherlands
| | | | - Wendy M. P. J. Sol
- Department of NephrologyLeiden University Medical CenterLeidenThe Netherlands
| | - Hetty de Boer
- Department of NephrologyLeiden University Medical CenterLeidenThe Netherlands
| | - Gangqi Wang
- Department of NephrologyLeiden University Medical CenterLeidenThe Netherlands
| | - Vanessa L. S. LaPointe
- MERLN Institute for Technology–Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Johan van der Vlag
- Department of NephrologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Cees van Kooten
- Department of NephrologyLeiden University Medical CenterLeidenThe Netherlands
| | | | - Melissa H. Little
- Department of NephrologyLeiden University Medical CenterLeidenThe Netherlands,Murdoch Childrens Research InstituteMelbourneAustralia,Department of PediatricsThe University of MelbourneMelbourneAustralia
| | - Marten A. Engelse
- Department of NephrologyLeiden University Medical CenterLeidenThe Netherlands
| | - Ton J. Rabelink
- Department of NephrologyLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
31
|
Koide H, Yoshimatsu K, Hoshino Y, Ariizumi S, Okishima A, Ide T, Egami H, Hamashima Y, Nishimura Y, Kanazawa H, Miura Y, Asai T, Oku N, Shea KJ. Sequestering and inhibiting a vascular endothelial growth factor in vivo by systemic administration of a synthetic polymer nanoparticle. J Control Release 2018; 295:13-20. [PMID: 30578808 DOI: 10.1016/j.jconrel.2018.12.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022]
Abstract
Protein affinity reagents (PARs), frequently antibodies, are essential tools for basic research, diagnostics, separations and for clinical applications. However, there is growing concern about the reproducibility, quality and cost of recombinant and animal-derived antibodies. This has prompted the development of alternatives that could offer economic, and time-saving advantages without the use of living organisms. Synthetic copolymer nanoparticles (NPs), engineered with affinity for specific protein targets, are potential alternatives to PARs. Although there are now a number of examples of abiotic protein affinity reagents (APARs), most have been evaluated in vitro limiting a realistic assessment of their potential for more demanding, practical in vivo applications. We demonstrate for the first time that an abiotic copolymer hydrogel nanoparticle (NP1) engineered to bind a key signaling protein, vascular endothelial growth factor (VEGF165), functions in vivo to suppress tumor growth by regulating angiogenesis. Lightly cross-linked N-isopropylacrylamide based NPs that incorporate both sulfated N-acetylglucosamine and hydrophobic monomers were optimized by dynamic chemical evolution for VEGF165 affinity. NP1 efficacy in vivo was evaluated by systemic administration to tumor-bearing mice. The study found that NP1 suppresses tumor growth and reduces tumor vasculature density. Combination therapy with doxorubicin resulted in increased doxorubicin concentration in the tumor and dramatic inhibition of tumor growth. NP1 treatment did not show off target anti-coagulant activity. In addition, >97% of injected NPs are rapidly excreted from the body following IV injection. These results establish the use of APARs as inhibitors of protein-protein interactions in vivo and may point the way to their broader use as abiotic, cost effective protein affinity reagents for the treatment of certain cancers and more broadly for regulating signal transduction.
Collapse
Affiliation(s)
- Hiroyuki Koide
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Keiichi Yoshimatsu
- Department of Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Yu Hoshino
- Department of Chemical Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Saki Ariizumi
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Anna Okishima
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Takafumi Ide
- Department of Synthetic Organic Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Hiromichi Egami
- Department of Synthetic Organic Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yoshitaka Hamashima
- Department of Synthetic Organic Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yuri Nishimura
- Department of Chemical Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Hiroaki Kanazawa
- Department of Functional Anatomy, School of Nursing, University of Shizuoka, 52-1 Yada, Shizuoka 422-8526, Japan
| | - Yoshiko Miura
- Department of Chemical Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Kenneth J Shea
- Department of Chemistry, University of California Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
32
|
Paderi J, Prestwich GD, Panitch A, Boone T, Stuart K. Glycan Therapeutics: Resurrecting an Almost Pharma‐Forgotten Drug Class. ADVANCED THERAPEUTICS 2018; 1. [DOI: 10.1002/adtp.201800082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Indexed: 01/04/2025]
Abstract
AbstractDespite their enormous potential, glycans as therapeutics yet remain a widely untapped drug class. This overview shares the viewpoint that glycans have been aptly termed the “dark matter” of biology and have thus been largely ignored for decades. Provided herein is a background on the multiple structures and functions of glycan therapeutics, and focuses on examples and case studies of the glycan therapeutics in clinical use or in a clinical development. Perspectives on various hurdles are also provided, such as regulatory or scientific messaging and how these can influence the clinical development of this drug category. Finally some of the necessary changes in perception, education, and research infrastructure for continued support and advancement of this promising category of therapeutics are described.
Collapse
Affiliation(s)
- John Paderi
- Symic Bio, Inc. 5980 Horton St. 94608 Emeryville CA USA
| | - Glenn D. Prestwich
- Symic Bio, Inc. 5980 Horton St. 94608 Emeryville CA USA
- Department of Medicinal Chemistry University of Utah 84112 Salt Lake City UT USA
- Washington State University Health Sciences Spokane 99210 Spokane WA USA
| | - Alyssa Panitch
- Symic Bio, Inc. 5980 Horton St. 94608 Emeryville CA USA
- University of California 95616 Davis CA USA
| | - Tom Boone
- Symic Bio, Inc. 5980 Horton St. 94608 Emeryville CA USA
| | - Kate Stuart
- Symic Bio, Inc. 5980 Horton St. 94608 Emeryville CA USA
| |
Collapse
|
33
|
Nih LR, Gojgini S, Carmichael ST, Segura T. Dual-function injectable angiogenic biomaterial for the repair of brain tissue following stroke. NATURE MATERIALS 2018; 17:642-651. [PMID: 29784996 PMCID: PMC6019573 DOI: 10.1038/s41563-018-0083-8] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 04/16/2018] [Indexed: 04/14/2023]
Abstract
Stroke is the primary cause of disability due to the brain's limited ability to regenerate damaged tissue. After stroke, an increased inflammatory and immune response coupled with severely limited angiogenesis and neuronal growth results in a stroke cavity devoid of normal brain tissue. In the adult, therapeutic angiogenic materials have been used to repair ischaemic tissues through the formation of vascular networks. However, whether a therapeutic angiogenic material can regenerate brain tissue and promote neural repair is poorly understood. Here we show that the delivery of an engineered immune-modulating angiogenic biomaterial directly to the stroke cavity promotes tissue formation de novo, and results in axonal networks along thee generated blood vessels. This regenerated tissue produces functional recovery through the established axonal networks. Thus, this biomaterials approach generates a vascularized network of regenerated functional neuronal connections within previously dead tissue and lays the groundwork for the use of angiogenic materials to repair other neurologically diseased tissues.
Collapse
Affiliation(s)
- Lina R Nih
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA, USA
- Department of Neurology David Geffen School of Medicine, University of California, Los Angeles, USA, CA
| | - Shiva Gojgini
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA, USA
| | - S Thomas Carmichael
- Department of Neurology David Geffen School of Medicine, University of California, Los Angeles, USA, CA.
| | - Tatiana Segura
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA, USA.
- Department of Biomedical Engineering, Neurology, Dermatology, Duke University, Durham, NC, USA.
| |
Collapse
|
34
|
Uciechowska-Kaczmarzyk U, Babik S, Zsila F, Bojarski KK, Beke-Somfai T, Samsonov SA. Molecular dynamics-based model of VEGF-A and its heparin interactions. J Mol Graph Model 2018; 82:157-166. [DOI: 10.1016/j.jmgm.2018.04.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 11/28/2022]
|
35
|
Peach CJ, Mignone VW, Arruda MA, Alcobia DC, Hill SJ, Kilpatrick LE, Woolard J. Molecular Pharmacology of VEGF-A Isoforms: Binding and Signalling at VEGFR2. Int J Mol Sci 2018; 19:E1264. [PMID: 29690653 PMCID: PMC5979509 DOI: 10.3390/ijms19041264] [Citation(s) in RCA: 329] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/14/2018] [Accepted: 04/16/2018] [Indexed: 02/07/2023] Open
Abstract
Vascular endothelial growth factor-A (VEGF-A) is a key mediator of angiogenesis, signalling via the class IV tyrosine kinase receptor family of VEGF Receptors (VEGFRs). Although VEGF-A ligands bind to both VEGFR1 and VEGFR2, they primarily signal via VEGFR2 leading to endothelial cell proliferation, survival, migration and vascular permeability. Distinct VEGF-A isoforms result from alternative splicing of the Vegfa gene at exon 8, resulting in VEGFxxxa or VEGFxxxb isoforms. Alternative splicing events at exons 5⁻7, in addition to recently identified posttranslational read-through events, produce VEGF-A isoforms that differ in their bioavailability and interaction with the co-receptor Neuropilin-1. This review explores the molecular pharmacology of VEGF-A isoforms at VEGFR2 in respect to ligand binding and downstream signalling. To understand how VEGF-A isoforms have distinct signalling despite similar affinities for VEGFR2, this review re-evaluates the typical classification of these isoforms relative to the prototypical, “pro-angiogenic” VEGF165a. We also examine the molecular mechanisms underpinning the regulation of VEGF-A isoform signalling and the importance of interactions with other membrane and extracellular matrix proteins. As approved therapeutics targeting the VEGF-A/VEGFR signalling axis largely lack long-term efficacy, understanding these isoform-specific mechanisms could aid future drug discovery efforts targeting VEGF receptor pharmacology.
Collapse
Affiliation(s)
- Chloe J Peach
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
| | - Viviane W Mignone
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
- CAPES-University of Nottingham Programme in Drug Discovery, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Maria Augusta Arruda
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
- CAPES-University of Nottingham Programme in Drug Discovery, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Diana C Alcobia
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
| | - Laura E Kilpatrick
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
| |
Collapse
|
36
|
Kamon Y, Takeuchi T. Molecularly Imprinted Nanocavities Capable of Ligand-Binding Domain and Size/Shape Recognition for Selective Discrimination of Vascular Endothelial Growth Factor Isoforms. ACS Sens 2018; 3:580-586. [PMID: 29441779 DOI: 10.1021/acssensors.7b00622] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Vascular endothelial growth factor 165 (VEGF165) is known to be predominantly expressed in the first stage of vascularization; therefore, the detection of VEGF165 is important in the stage diagnosis of cancers. Molecularly imprinted nanocavities, capable of the selective discrimination of VEGF165 from other VEGF isoforms, were prepared by surface-initiated atom transfer radical polymerization. VEGF165 was immobilized on a gold-coated glass substrate by anchored heparin moieties, where the immobilized heparin was able to capture VEGF165 by binding with the heparin-binding domain (HBD) on VEGF165. Molecular imprinting was conducted on the immobilized VEGF165 by using methacrylic acid (MAA) as a functional monomer to interact with basic amino acids outside of the HBD of VEGF165 by electrostatic interaction. After the removal of VEGF165 from the obtained polymer thin layer (ca. 7 nm), VEGF165-imprinted nanocavities remained, in which the heparin moiety and MAA residues were located in suitable positions for VEGF165 recognition. The molecularly imprinted polymer (MIP) thin layer showed a binding affinity for VEGF165 (dissociation constant: 3.4 nM) that was ten times higher than that of the substrate before polymerization (heparin-immobilized substrate). A much lower binding affinity for VEGF121, which contains no heparin-binding domain, was observed. Moreover, the MIP thin layer distinguished VEGF165 from VEGF189, which possesses a larger molecular size than VEGF165, an amino acid sequence homology of 87%, and contains HBDs, whereas the heparin-immobilized substrate showed almost no selectivity. These results suggested that the heparin moiety within the nanocavity provided HBD selectivity and the polymer matrix composed of the molecularly imprinted nanocavity provided size/shape selectivity, which resulted in the highly selective discrimination of VEGF isoforms.
Collapse
Affiliation(s)
- Yuri Kamon
- Graduate School of Engineering, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Toshifumi Takeuchi
- Graduate School of Engineering, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| |
Collapse
|
37
|
Varongchayakul N, Huttner D, Grinstaff MW, Meller A. Sensing Native Protein Solution Structures Using a Solid-state Nanopore: Unraveling the States of VEGF. Sci Rep 2018; 8:1017. [PMID: 29343861 PMCID: PMC5772516 DOI: 10.1038/s41598-018-19332-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/27/2017] [Indexed: 01/01/2023] Open
Abstract
Monitoring individual proteins in solution while simultaneously obtaining tertiary and quaternary structural information is challenging. In this study, translocation of the vascular endothelial growth factor (VEGF) protein through a solid-state nanopore (ssNP) produces distinct ion-current blockade amplitude levels and durations likely corresponding to monomer, dimer, and higher oligomeric states. Upon changing from a non-reducing to a reducing condition, ion-current blockage events from the monomeric state dominate, consistent with the expected reduction of the two inter-chain VEGF disulfide bonds. Cleavage by plasmin and application of either a positive or a negative NP bias results in nanopore signals corresponding either to the VEGF receptor recognition domain or to the heparin binding domain, accordingly. Interestingly, multi-level analysis of VEGF events reveals how individual domains affect their translocation pattern. Our study shows that careful characterization of ssNP results elucidates real-time structural information about the protein, thereby complementing classical techniques for structural analysis of proteins in solution with the added advantage of quantitative single-molecule resolution of native proteins.
Collapse
Affiliation(s)
- Nitinun Varongchayakul
- Departments of Biomedical Engineering, Boston University, Boston, 02215, Massachusetts, USA
| | - Diana Huttner
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| | - Mark W Grinstaff
- Departments of Biomedical Engineering, Boston University, Boston, 02215, Massachusetts, USA. .,Department of Chemistry, Boston University, Boston, 02215, Massachusetts, USA. .,School of Medicine, Boston University, Boston, 02215, Massachusetts, USA.
| | - Amit Meller
- Departments of Biomedical Engineering, Boston University, Boston, 02215, Massachusetts, USA. .,Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, 32000, Israel.
| |
Collapse
|
38
|
Abstract
All known splice isoforms of vascular endothelial growth factor A (VEGF-A) can bind to the receptor tyrosine kinases VEGFR-1 and VEGFR-2. We focus here on VEGF-A121a and VEGF-A165a, two of the most abundant VEGF-A splice isoforms in human tissue 1 , and their ability to bind the Neuropilin co-receptors NRP1 and NRP2. The Neuropilins are key vascular, immune, and nervous system receptors on endothelial cells, neuronal axons, and regulatory T cells respectively. They serve as co-receptors for the Plexins in Semaphorin binding on neuronal and vascular endothelial cells, and for the VEGFRs in VEGF binding on vascular and lymphatic endothelial cells, and thus regulate the initiation and coordination of cell signaling by Semaphorins and VEGFs. 2 There is conflicting evidence in the literature as to whether only heparin-binding VEGF-A isoforms - that is, isoforms with domains encoded by exons 6 and/or 7 plus 8a - bind to Neuropilins on endothelial cells. While it is clear that VEGF-A165a binds to both NRP1 and NRP2, published studies do not all agree on the ability of VEGF-A121a to bind NRPs. Here, we review and attempt to reconcile evidence for and against VEGF-A121a binding to Neuropilins. This evidence suggests that, in vitro, VEGF-A121a can bind to both NRP1 and NRP2 via domains encoded by exons 5 and 8a; in the case of NRP1, VEGF-A121a binds with lower affinity than VEGF-A165a. In in vitro cell culture experiments, both NRP1 and NRP2 can enhance VEGF-A121a-induced phosphorylation of VEGFR2 and downstream signaling including proliferation. However, unlike VEGFA-165a, experiments have shown that VEGF-A121a does not 'bridge' VEGFR2 and NRP1, i.e. it does not bind both receptors simultaneously at their extracellular domain. Thus, the mechanism by which Neuropilins potentiate VEGF-A121a-mediated VEGFR2 signaling may be different from that for VEGF-A165a. We suggest such an alternate mechanism: interactions between NRP1 and VEGFR2 transmembrane (TM) and intracellular (IC) domains.
Collapse
Affiliation(s)
- Sarvenaz Sarabipour
- a Institute for Computational Medicine, Department of Biomedical Engineering, Institute for NanoBioTechnology , Johns Hopkins University , Baltimore , MD , USA
| | - Feilim Mac Gabhann
- a Institute for Computational Medicine, Department of Biomedical Engineering, Institute for NanoBioTechnology , Johns Hopkins University , Baltimore , MD , USA
| |
Collapse
|
39
|
Contribution of vascular endothelial growth factor receptor-2 sialylation to the process of angiogenesis. Oncogene 2017; 36:6531-6541. [PMID: 28783175 DOI: 10.1038/onc.2017.243] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 05/30/2017] [Accepted: 06/12/2017] [Indexed: 01/18/2023]
Abstract
Vascular endothelial growth factor receptor-2 (VEGFR2) is the main pro-angiogenic receptor expressed by endothelial cells (ECs). Using surface plasmon resonance, immunoprecipitation, enzymatic digestion, immunofluorescence and cross-linking experiments with specific sugar-binding lectins, we demonstrated that VEGFR2 bears both α,1-fucose and α(2,6)-linked sialic acid (NeuAc). However, only the latter is required for VEGF binding to VEGFR2 and consequent VEGF-dependent VEGFR2 activation and motogenic response in ECs. Notably, downregulation of β-galactoside α(2,6)-sialyltransferase expression by short hairpin RNA transduction inhibits VEGFR2 α(2,6) sialylation that is paralleled by an increase of β-galactoside α(2,3)-sialyltransferase expression. This results in an ex-novo α(2,3)-NeuAc sialylation of the receptor that functionally replaces the lacking α(2,6)-NeuAc, thus allowing VEGF/VEGFR2 interaction. In keeping with the role of VEGFR2 sialylation in angiogenesis, the α(2,6)-NeuAc-binding lectin Sambucus nigra (SNA) prevents VEGF-dependent VEGFR2 autophosphorylation and EC motility, proliferation and motogenesis. In addition, SNA exerts a VEGF-antagonist activity in tridimensional angiogenesis models in vitro and in the chick-embryo chorioallantoic membrane neovascularization assay and mouse matrigel plug assay in vivo. In conclusion, VEGFR2-associated NeuAc plays an important role in modulating VEGF/VEGFR2 interaction, EC pro-angiogenic activation and neovessel formation. VEGFR2 sialylation may represent a target for the treatment of angiogenesis-dependent diseases.
Collapse
|
40
|
Pacelli S, Acosta F, Chakravarti AR, Samanta SG, Whitlow J, Modaresi S, Ahmed RPH, Rajasingh J, Paul A. Nanodiamond-based injectable hydrogel for sustained growth factor release: Preparation, characterization and in vitro analysis. Acta Biomater 2017; 58:479-491. [PMID: 28532899 PMCID: PMC5560430 DOI: 10.1016/j.actbio.2017.05.026] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/06/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022]
Abstract
Nanodiamonds (NDs) represent an emerging class of carbon nanomaterials that possess favorable physical and chemical properties to be used as multifunctional carriers for a variety of bioactive molecules. Here we report the synthesis and characterization of a new injectable ND-based nanocomposite hydrogel which facilitates a controlled release of therapeutic molecules for regenerative applications. In particular, we have formulated a thermosensitive hydrogel using gelatin, chitosan and NDs that provides a sustained release of exogenous human vascular endothelial growth factor (VEGF) for wound healing applications. Addition of NDs improved the mechanical properties of the injectable hydrogels without affecting its thermosensitive gelation properties. Biocompatibility of the generated hydrogel was verified by in vitro assessment of apoptotic gene expressions and anti-inflammatory interleukin productions. NDs were complexed with VEGF and the inclusion of this complex in the hydrogel network enabled the sustained release of the angiogenic growth factor. These results suggest for the first time that NDs can be used to formulate a biocompatible, thermosensitive and multifunctional hydrogel platform that can function both as a filling agent to modulate hydrogel properties, as well as a delivery platform for the controlled release of bioactive molecules and growth factors. STATEMENT OF SIGNIFICANCE One of the major drawbacks associated with the use of conventional hydrogels as carriers of growth factors is their inability to control the release kinetics of the loaded molecules. In fact, in most cases, a burst release is inevitable leading to diminished therapeutic effects and unsuccessful therapies. As a potential solution to this issue, we hereby propose a strategy of incorporating ND complexes within an injectable hydrogel matrix. The functional groups on the surface of the NDs can establish interactions with the model growth factor VEGF and promote a prolonged release from the polymer network, therefore, providing a longer therapeutic effect. Our strategy demonstrates the efficacy of using NDs as an essential component for the design of a novel injectable nanocomposite system with improved release capabilities.
Collapse
Affiliation(s)
- Settimio Pacelli
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, KS, United States
| | - Francisca Acosta
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, KS, United States
| | - Aparna R Chakravarti
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, KS, United States
| | - Saheli G Samanta
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jonathan Whitlow
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, KS, United States
| | - Saman Modaresi
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, KS, United States
| | - Rafeeq P H Ahmed
- Department of Pathology, University of Cincinnati, 231-Albert Sabin Way, Cincinnati 45267, United States
| | - Johnson Rajasingh
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Arghya Paul
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, KS, United States.
| |
Collapse
|
41
|
Lai PX, Chen CW, Wei SC, Lin TY, Jian HJ, Lai IPJ, Mao JY, Hsu PH, Lin HJ, Tzou WS, Chen SY, Harroun SG, Lai JY, Huang CC. Ultrastrong trapping of VEGF by graphene oxide: Anti-angiogenesis application. Biomaterials 2016; 109:12-22. [DOI: 10.1016/j.biomaterials.2016.09.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/31/2016] [Accepted: 09/07/2016] [Indexed: 01/13/2023]
|
42
|
Zaman P, Wang J, Blau A, Wang W, Li T, Kohane DS, Loscalzo J, Zhang YY. Incorporation of heparin-binding proteins into preformed dextran sulfate-chitosan nanoparticles. Int J Nanomedicine 2016; 11:6149-6159. [PMID: 27920522 PMCID: PMC5125769 DOI: 10.2147/ijn.s119174] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Incorporation of proteins into dextran sulfate (DS)-chitosan (CS) nanoparticles (DSCS NPs) is commonly performed using entrapment procedures, in which protein molecules are mixed with DS and CS until particle formation occurs. As DS is an analog of heparin, the authors examined whether proteins could be directly incorporated into preformed DSCS NPs through a heparin binding domain-mediated interaction. The authors formulated negatively-charged DSCS NPs, and quantified the amount of charged DS in the outer shell of the particles. The authors then mixed the DSCS NPs with heparin-binding proteins (SDF-1α, VEGF, FGF-2, BMP-2, or lysozyme) to achieve incorporation. Data show that for DSCS NPs containing 100 nmol charged glucose sulfate units in DS, up to ~1.5 nmol of monomeric or ~0.75 nmol of dimeric heparin-binding proteins were incorporated without significantly altering the size or zeta potential of the particles. Incorporation efficiencies of these proteins were 95%–100%. In contrast, serum albumin or serum globulin showed minimal incorporation (8% and 4%, respectively) in 50% physiological saline, despite their large adsorption in water (80% and 92%, respectively). The NP-incorporated SDF-1α and VEGF exhibited full activity and sustained thermal stability. An in vivo aerosolization study showed that NP-incorporated SDF-1α persisted in rat lungs for 72 h (~34% remaining), while free SDF-1α was no longer detectable after 16 h. As many growth factors and cytokines contain heparin-binding sites/domains, incorporation into preformed DSCS NPs could facilitate in vivo applications of these proteins.
Collapse
Affiliation(s)
- Paula Zaman
- Department of Medicine, Brigham and Women's Hospital
| | - Julia Wang
- Department of Medicine, Brigham and Women's Hospital
| | - Adam Blau
- Department of Medicine, Brigham and Women's Hospital
| | - Weiping Wang
- Department of Anesthesiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tina Li
- Department of Medicine, Brigham and Women's Hospital
| | - Daniel S Kohane
- Department of Anesthesiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Ying-Yi Zhang
- Department of Medicine, Brigham and Women's Hospital
| |
Collapse
|
43
|
Riederer MS, Requist BD, Payne KA, Way JD, Krebs MD. Injectable and microporous scaffold of densely-packed, growth factor-encapsulating chitosan microgels. Carbohydr Polym 2016; 152:792-801. [PMID: 27516331 PMCID: PMC4988396 DOI: 10.1016/j.carbpol.2016.07.052] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 12/20/2022]
Abstract
In this work, an emulsion crosslinking method was developed to produce chitosan-genipin microgels which acted as an injectable and microporous scaffold. Chitosan was characterized with respect to pH by light scattering and aqueous titration. Microgels were characterized with swelling, light scattering, and rheometry of densely-packed microgel solutions. The results suggest that as chitosan becomes increasingly deprotonated above the pKa, repulsive forces diminish and intermolecular attractions cause pH-responsive chain aggregation; leading to microgel-microgel aggregation as well. The microgels with the most chitosan and least cross-linker showed the highest yield stress and a storage modulus of 16kPa when condensed as a microgel paste at pH 7.4. Two oppositely-charged growth factors could be encapsulated into the microgels and endothelial cells were able to proliferate into the 3D microgel scaffold. This work motivates further research on the applications of the chitosan microgel scaffold as an injectable and microporous scaffold in regenerative medicine.
Collapse
Affiliation(s)
- Michael S Riederer
- Department of Chemical & Biological Engineering, Colorado School of Mines, 1613 Illinois Street, Golden, CO 80401, United States.
| | - Brennan D Requist
- Department of Chemical & Biological Engineering, Colorado School of Mines, 1613 Illinois Street, Golden, CO 80401, United States.
| | - Karin A Payne
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, 12800 E. 19th Avenue, Aurora, CO 80045, United States.
| | - J Douglas Way
- Department of Chemical & Biological Engineering, Colorado School of Mines, 1613 Illinois Street, Golden, CO 80401, United States.
| | - Melissa D Krebs
- Department of Chemical & Biological Engineering, Colorado School of Mines, 1613 Illinois Street, Golden, CO 80401, United States.
| |
Collapse
|
44
|
Uzunalli G, Mammadov R, Yesildal F, Alhan D, Ozturk S, Ozgurtas T, Guler MO, Tekinay AB. Angiogenic Heparin-Mimetic Peptide Nanofiber Gel Improves Regenerative Healing of Acute Wounds. ACS Biomater Sci Eng 2016; 3:1296-1303. [DOI: 10.1021/acsbiomaterials.6b00165] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Gozde Uzunalli
- Institute
of Materials Science and Nanotechnology, National Nanotechnology Research
Center (UNAM), Bilkent University, Ankara, Turkey 06800
| | - Rashad Mammadov
- Institute
of Materials Science and Nanotechnology, National Nanotechnology Research
Center (UNAM), Bilkent University, Ankara, Turkey 06800
| | - Fatih Yesildal
- Department
of Medical Biochemistry, Diyarbakir Military Hospital, Diyarbakir, Turkey
| | - Dogan Alhan
- Gulhane Military Medical Academy, Ankara, Turkey
| | | | | | - Mustafa O. Guler
- Institute
of Materials Science and Nanotechnology, National Nanotechnology Research
Center (UNAM), Bilkent University, Ankara, Turkey 06800
| | - Ayse B. Tekinay
- Institute
of Materials Science and Nanotechnology, National Nanotechnology Research
Center (UNAM), Bilkent University, Ankara, Turkey 06800
| |
Collapse
|
45
|
Tsai YCI, Fotinou C, Rana R, Yelland T, Frankel P, Zachary I, Djordjevic S. Structural studies of neuropilin-2 reveal a zinc ion binding site remote from the vascular endothelial growth factor binding pocket. FEBS J 2016; 283:1921-34. [PMID: 26991001 PMCID: PMC4914954 DOI: 10.1111/febs.13711] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/24/2016] [Accepted: 03/10/2016] [Indexed: 01/08/2023]
Abstract
Neuropilin-2 is a transmembrane receptor involved in lymphangiogenesis and neuronal development. In adults, neuropilin-2 and its homologous protein neuropilin-1 have been implicated in cancers and infection. Molecular determinants of the ligand selectivity of neuropilins are poorly understood. We have identified and structurally characterized a zinc ion binding site on human neuropilin-2. The neuropilin-2-specific zinc ion binding site is located near the interface between domains b1 and b2 in the ectopic region of the protein, remote from the neuropilin binding site for its physiological ligand, i.e. vascular endothelial growth factor. We also present an X-ray crystal structure of the neuropilin-2 b1 domain in a complex with the C-terminal sub-domain of VEGF-A. Zn(2+) binding to neuropilin-2 destabilizes the protein structure but this effect was counteracted by heparin, suggesting that modifications by glycans and zinc in the extracellular matrix may affect functional neuropilin-2 ligand binding and signalling activity.
Collapse
Affiliation(s)
- Yi-Chun Isabella Tsai
- Institute of Structural and Molecular Biology, University College London, London, UK
| | | | - Rohini Rana
- Magnus Life Science, University College London, London, UK
| | - Tamas Yelland
- Institute of Structural and Molecular Biology, University College London, London, UK
| | - Paul Frankel
- Centre for Cardiovascular Biology & Medicine, BHF Laboratories at University College London, London, UK
| | - Ian Zachary
- Centre for Cardiovascular Biology & Medicine, BHF Laboratories at University College London, London, UK
| | - Snezana Djordjevic
- Institute of Structural and Molecular Biology, University College London, London, UK
| |
Collapse
|
46
|
Xiong GM, Yap YZ, Choong C. Single-step synthesis of heparin-doped polypyrrole nanoparticles for delivery of angiogenic factor. Nanomedicine (Lond) 2016; 11:749-65. [DOI: 10.2217/nnm.16.13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim: To perform one-pot synthesis of heparin-immobilized polypyrrole (PPy) nanoparticles and evaluate the use of these nanoparticles for the delivery of VEGF. Materials & methods: Heparin-stabilized synthesis of PPy nanoparticles was performed via oxidative polymerization. VEGF-bound PPy-heparin nanoparticles were delivered to endothelial cells and bioactivity of VEGF was assessed by Matrigel tube formation. Results: Size-controllable synthesis of heparin-doped PPy nanoparticles was achieved, and heparin promoted the conjugation of VEGF. Angiogenic activity of the VEGF-conjugated PPy nanoparticles was verified. Conclusion: Heparin-doped PPy nanoparticles can be synthesized using one-pot reaction and provide a delivery platform by which VEGF can be conjugated onto.
Collapse
Affiliation(s)
- Gordon M Xiong
- School of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Yi Zhen Yap
- School of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Cleo Choong
- School of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
- KK Research Centre, KK Women's & Children Hospital, 100 Bukit Timah Road, 229899 Singapore
| |
Collapse
|
47
|
Jin K, Li B, Lou L, Xu Y, Ye X, Yao K, Ye J, Gao C. In vivo vascularization of MSC-loaded porous hydroxyapatite constructs coated with VEGF-functionalized collagen/heparin multilayers. Sci Rep 2016; 6:19871. [PMID: 26794266 PMCID: PMC4726420 DOI: 10.1038/srep19871] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 12/18/2015] [Indexed: 12/18/2022] Open
Abstract
Rapid and adequate vascularization is vital to the long-term success of porous orbital enucleation implants. In this study, porous hydroxyapatite (HA) scaffolds coated with vascular endothelial growth factor (VEGF)-functionalized collagen (COL)/heparin (HEP) multilayers (porosity 75%, pore size 316.8 ± 77.1 μm, VEGF dose 3.39 ng/mm3) were fabricated to enhance vascularization by inducing the differentiation of mesenchymal stem cells (MSCs) to endothelial cells. The in vitro immunofluorescence staining, quantitative real-time polymerase chain reaction (qRT-PCR), and western blotting results demonstrated that the expression of the endothelial differentiation markers CD31, Flk-1, and von Willebrand factor (vWF) was significantly increased in the HA/(COL/HEP)5/VEGF/MSCs group compared with the HA/VEGF/MSCs group. Moreover, the HA/(COL/HEP)5 scaffolds showed a better entrapment of the MSCs and accelerated cell proliferation. The in vivo assays showed that the number of newly formed vessels within the constructs after 28 d was significantly higher in the HA/(COL/HEP)5/VEGF/MSCs group (51.9 ± 6.3/mm2) than in the HA (26.7 ± 2.3/mm2) and HA/VEGF/MSCs (38.2 ± 2.4/mm2) groups. The qRT-PCR and western blotting results demonstrated that the HA/(COL/HEP)5/VEGF/MSCs group also had the highest expression of CD31, Flk-1, and vWF at both the mRNA and protein levels.
Collapse
Affiliation(s)
- Kai Jin
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Bo Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Lixia Lou
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Yufeng Xu
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Xin Ye
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Ke Yao
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Juan Ye
- Department of Ophthalmology, the Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
48
|
Walker A, Chung CW, Neu M, Burman M, Batuwangala T, Jones G, Tang CM, Steward M, Mullin M, Tournier N, Lewis A, Korczynska J, Chung V, Catchpole I. Novel Interaction Mechanism of a Domain Antibody-based Inhibitor of Human Vascular Endothelial Growth Factor with Greater Potency than Ranibizumab and Bevacizumab and Improved Capacity over Aflibercept. J Biol Chem 2016; 291:5500-5511. [PMID: 26728464 PMCID: PMC4786692 DOI: 10.1074/jbc.m115.691162] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Indexed: 12/27/2022] Open
Abstract
A potent VEGF inhibitor with novel antibody architecture and antigen binding mode has been developed. The molecule, hereafter referred to as VEGF dual dAb (domain antibody), was evaluated in vitro for binding to VEGF and for potency in VEGF-driven models and compared with other anti-VEGF biologics that have been used in ocular anti-angiogenic therapeutic regimes. VEGF dual dAb is more potent than bevacizumab and ranibizumab for VEGF binding, inhibition of VEGF receptor binding assays (RBAs), and VEGF-driven in vitro models of angiogenesis and displays comparable inhibition to aflibercept (Eylea). VEGF dual dAb is dimeric, and each monomer contains two distinct anti-VEGF domain antibodies attached via linkers to a human IgG1 Fc domain. Mechanistically, the enhanced in vitro potency of VEGF dual dAb, in comparison to other anti-VEGF biologics, can be explained by increased binding stoichiometry. A consistent model of the target engagement has been built based on the x-ray complexes of each of the two isolated domain antibodies with the VEGF antigen.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Vicky Chung
- BioPharm Process Research, GSK Medicine's Research Centre, Stevenage, Herts SG1 2NY, United Kingdom
| | | |
Collapse
|
49
|
Azhar A, Khan MS, Swaminathan A, Naseem A, Chatterjee S, Jairajpuri MA. Oxidized antithrombin is a dual inhibitor of coagulation and angiogenesis: Importance of low heparin affinity. Int J Biol Macromol 2016; 82:541-50. [DOI: 10.1016/j.ijbiomac.2015.10.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/01/2015] [Accepted: 10/05/2015] [Indexed: 01/09/2023]
|
50
|
Azevedo HS, Pashkuleva I. Biomimetic supramolecular designs for the controlled release of growth factors in bone regeneration. Adv Drug Deliv Rev 2015; 94:63-76. [PMID: 26325686 DOI: 10.1016/j.addr.2015.08.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 08/17/2015] [Accepted: 08/25/2015] [Indexed: 12/13/2022]
Abstract
The extracellular matrix (ECM) of tissues is an assembly of insoluble macromolecules that specifically interact with soluble bioactive molecules and regulate their distribution and availability to cells. Recapitulating this ability has been an important target in controlled growth factor delivery strategies for tissue regeneration and requires the design of multifunctional carriers. This review describes the integration of supramolecular interactions on the design of delivery strategies that encompass self-assembling and engineered affinity components to construct advanced biomimetic carriers for growth factor delivery. Several glycan- and peptide-based self-assemblies reported in the literature are highlighted and commented upon. These examples demonstrate how molecular design and chemistry are successfully employed to create versatile multifunctional molecules which self-assemble/disassemble in a precisely predicted manner, thus controlling compartmentalization, transport and delivery. Finally, we discuss whether recent advances in the design and preparation of supramolecular delivery systems have been sufficient to drive real translation towards a clinical impact.
Collapse
Affiliation(s)
- Helena S Azevedo
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK; Institute of Bioengineering, Queen Mary University of London, Mile End Road, London E1 4NS, UK.
| | - Iva Pashkuleva
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|