1
|
Zhu S, Song Z, Tapayan AS, Singh K, Wang KW, Chien Hagar HT, Zhang J, Kim H, Thepsuwan P, Kuo MH, Zhang K, Nguyen HM. Effects of Heparan Sulfate Trisaccharide Containing Oleanolic Acid in Attenuating Hyperphosphorylated Tau-Induced Cell Dysfunction Associated with Alzheimer's Disease. J Med Chem 2025; 68:3356-3372. [PMID: 39842821 DOI: 10.1021/acs.jmedchem.4c02563] [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: 01/24/2025]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, marked by progressive brain degeneration and cognitive decline. A major pathological feature of AD is the accumulation of hyperphosphorylated tau (p-tau) in the form of neurofibrillary tangles (NFTs), which leads to neuronal death and neurodegeneration. P-tau also induces endoplasmic reticulum (ER) stress and activates the unfolded protein response, causing inflammation and apoptosis. Additionally, p-tau spreads in the brain through interactions with heparan sulfate (HS) proteoglycans, promoting aggregation and internalization. Targeting the tau-HS interaction offers a potential therapeutic strategy for AD. We present a novel HS mimetic with a lipophilic oleanolic acid linker and a sulfated trisaccharide, which shows strong cytoprotective effects against p-tau. Moreover, this compound alleviates p-tau-induced ER stress and inflammation. Molecular docking studies indicate that the conjugation of oleanolic acid enhances binding between the ligand and tau protofilament cores, facilitating protective interactions. These findings provide a foundation for the development of novel HS mimetics, enabling further investigation of tau-HS interactions in AD and other tauopathies.
Collapse
Affiliation(s)
- Sanyong Zhu
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Zhenfeng Song
- Center for Molecular Medicine and Genetics, Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan 48202, United States
| | - April Sweet Tapayan
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Kartikey Singh
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Kuang-Wei Wang
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Hsiao-Tien Chien Hagar
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jicheng Zhang
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Hyunbae Kim
- Center for Molecular Medicine and Genetics, Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan 48202, United States
| | - Patty Thepsuwan
- Center for Molecular Medicine and Genetics, Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan 48202, United States
| | - Min-Hao Kuo
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan 48202, United States
| | - Hien M Nguyen
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| |
Collapse
|
2
|
Piszczatowski RT, Bülow HE, Steidl U. Heparan sulfates and heparan sulfate proteoglycans in hematopoiesis. Blood 2024; 143:2571-2587. [PMID: 38639475 PMCID: PMC11830984 DOI: 10.1182/blood.2023022736] [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: 10/04/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
ABSTRACT From signaling mediators in stem cells to markers of differentiation and lineage commitment to facilitators for the entry of viruses, such as HIV-1, cell surface heparan sulfate (HS) glycans with distinct modification patterns play important roles in hematopoietic biology. In this review, we provide an overview of the importance of HS and the proteoglycans (HSPGs) to which they are attached within the major cellular subtypes of the hematopoietic system. We summarize the roles of HSPGs, HS, and HS modifications within each main hematopoietic cell lineage of both myeloid and lymphoid arms. Lastly, we discuss the biological advances in the detection of HS modifications and their potential to further discriminate cell types within hematopoietic tissue.
Collapse
Affiliation(s)
- Richard T. Piszczatowski
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
- Department of Pediatrics, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY
| | - Hannes E. Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY
- Departments of Oncology, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY
- Blood Cancer Institute, Albert Einstein College of Medicine, Bronx, NY
- Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
3
|
Sun L, Chopra P, Tomris I, van der Woude R, Liu L, de Vries RP, Boons GJ. Well-Defined Heparin Mimetics Can Inhibit Binding of the Trimeric Spike of SARS-CoV-2 in a Length-Dependent Manner. JACS AU 2023; 3:1185-1195. [PMID: 37101566 PMCID: PMC10089289 DOI: 10.1021/jacsau.3c00042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 06/19/2023]
Abstract
The emergence of new SARS-CoV-2 variants and the dangers of long-covid necessitate the development of broad-acting therapeutics that can reduce viral burden. SARS-CoV-2 employs heparan sulfate (HS) as an initial cellular attachment factor, and therefore, there is interest in developing heparin as a therapeutic for SARS-CoV-2. Its use is, however, complicated by structural heterogeneity and the risk of causing bleeding and thrombocytopenia. Here, we describe the preparation of well-defined heparin mimetics by a controlled head-to-tail assembly of HS oligosaccharides having an alkyne or azide moiety by copper-catalyzed azide-alkyne cycloaddition (CuAAC). Alkyne- and azide-containing sulfated oligosaccharides were prepared from a common precursor by modifying an anomeric linker with 4-pentynoic acid and by enzymatic extension with an N-acetyl-glucosamine having an azide moiety at C-6 (GlcNAc6N3), respectively, followed by CuAAC. The process of enzymatic extension with GlcNAc6N3 followed by CuAAC with the desired alkyne-containing oligosaccharides could be repeated to give compounds composed of 20 and 27 monosaccharides, respectively. The heparin mimetics could inhibit the binding of the SARS-CoV-2 spike or RBD to immobilized heparin or to Vero E6 cells. The inhibitory potency increased with increasing chain length, and a compound composed of four sulfated hexasaccharides linked by triazoles had a similar potency as unfractionated heparin. Sequence analysis and HS microarray binding studies with a wide range of RBDs of variants of concern indicate that they have maintained HS-binding capabilities and selectivities. The heparin mimetics exhibit no or reduced binding to antithrombin-III and platelet factor 4, respectively, which are associated with side effects.
Collapse
Affiliation(s)
- Lifeng Sun
- Department
of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Pradeep Chopra
- Complex
Carbohydrate Research Center, The University
of Georgia, Athens, Georgia 30602, United States
| | - Ilhan Tomris
- Department
of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Roosmarijn van der Woude
- Department
of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Lin Liu
- Complex
Carbohydrate Research Center, The University
of Georgia, Athens, Georgia 30602, United States
| | - Robert P. de Vries
- Department
of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Geert-Jan Boons
- Department
of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
- Complex
Carbohydrate Research Center, The University
of Georgia, Athens, Georgia 30602, United States
- Bijvoet
Center for Biomolecular Research, Utrecht
University, 3584 CG Utrecht, The Netherlands
- Chemistry
Department, The University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
4
|
Yang C, Deng Y, Wang Y, Xia C, Mehta AY, Baker KJ, Samal A, Booneimsri P, Lertmaneedang C, Hwang S, Flynn JP, Cao M, Liu C, Zhu AC, Cummings RD, Lin C, Mohanty U, Niu J. Heparan sulfate glycomimetics via iterative assembly of "clickable" disaccharides. Chem Sci 2023; 14:3514-3522. [PMID: 37006675 PMCID: PMC10055906 DOI: 10.1039/d3sc00260h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 02/27/2023] [Indexed: 03/04/2023] Open
Abstract
Heparan sulfate (HS) glycosaminoglycans are widely expressed on the mammalian cell surfaces and extracellular matrices and play important roles in a variety of cell functions. Studies on the structure-activity relationships of HS have long been hampered by the challenges in obtaining chemically defined HS structures with unique sulfation patterns. Here, we report a new approach to HS glycomimetics based on iterative assembly of clickable disaccharide building blocks that mimic the disaccharide repeating units of native HS. Variably sulfated clickable disaccharides were facilely assembled into a library of mass spec-sequenceable HS-mimetic oligomers with defined sulfation patterns by solution-phase iterative syntheses. Microarray and surface plasmon resonance (SPR) binding assays corroborated molecular dynamics (MD) simulations and confirmed that these HS-mimetic oligomers bind protein fibroblast growth factor 2 (FGF2) in a sulfation-dependent manner consistent with that of the native HS. This work established a general approach to HS glycomimetics that can potentially serve as alternatives to native HS in both fundamental research and disease models.
Collapse
Affiliation(s)
- Cangjie Yang
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
| | - Yu Deng
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
| | - Yang Wang
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
| | - Chaoshuang Xia
- Center for Biomedical Mass Spectrometry, Boston University School of Medicine Boston MA 02118 USA
| | - Akul Y Mehta
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School Brookline Ave Boston MA 02215 USA
| | - Kelly J Baker
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School Brookline Ave Boston MA 02215 USA
| | - Anuj Samal
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
| | - Putthipong Booneimsri
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
- Department of Chemistry, Faculty of Science, Chulalongkorn University Bangkok 10330 Thailand
| | - Chanthakarn Lertmaneedang
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
- Department of Chemistry, Faculty of Science, Chulalongkorn University Bangkok 10330 Thailand
| | - Seung Hwang
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
| | - James P Flynn
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
| | - Muqing Cao
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
| | - Chao Liu
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
| | - Alec C Zhu
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School Brookline Ave Boston MA 02215 USA
| | - Cheng Lin
- Center for Biomedical Mass Spectrometry, Boston University School of Medicine Boston MA 02118 USA
| | - Udayan Mohanty
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
| | - Jia Niu
- Department of Chemistry, Boston College Chestnut Hill Massachusetts 02467 USA
| |
Collapse
|
5
|
Loka RS, Song Z, Sletten ET, Kayal Y, Vlodavsky I, Zhang K, Nguyen HM. Heparan Sulfate Mimicking Glycopolymer Prevents Pancreatic β Cell Destruction and Suppresses Inflammatory Cytokine Expression in Islets under the Challenge of Upregulated Heparanase. ACS Chem Biol 2022; 17:1387-1400. [PMID: 35658404 PMCID: PMC9251817 DOI: 10.1021/acschembio.1c00908] [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] [Indexed: 11/28/2022]
Abstract
Diabetes is a chronic disease in which the levels of blood glucose are too high because the body does not effectively produce insulin to meet its needs or is resistant to insulin. β Cells in human pancreatic islets produce insulin, which signals glucogen production by the liver and causes muscles and fat to uptake glucose. Progressive loss of insulin-producing β cells is the main cause of both type 1 and type 2 diabetes. Heparan sulfate (HS) is a ubiquitous polysaccharide found at the cell surface and in the extracellular matrix (ECM) of a variety of tissues. HS binds to and assembles proteins in ECM, thus playing important roles in the integrity of ECM (particularly basement membrane), barrier function, and ECM-cell interactions. Islet HS is highly expressed by the pancreatic β cells and critical for the survival of β cells. Heparanase is an endoglycosidase and cleaves islet HS in the pancreas, resulting in β-cell death and oxidative stress. Heparanase could also accelerate β-cell death by promoting cytokine release from ECM and secretion by activated inflammatory and endothelial cells. We demonstrate that HS-mimicking glycopolymer, a potent heparanase inhibitor, improves the survival of cultured mouse pancreatic β cells and protects HS contents under the challenge of heparanase in human pancreatic islets. Moreover, this HS-mimicking glycopolymer reduces the expression levels of cytokines (IL8, IL1β, and TNFα) and the gene encoding Toll-like Receptor 2 (TLR2) in human pancreatic islets.
Collapse
Affiliation(s)
- Ravi S Loka
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Zhenfeng Song
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Eric T Sletten
- Department of Chemistry, University of Iowa, Iowa City, Iowa 52242, United States
| | - Yasmin Kayal
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Hien M Nguyen
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| |
Collapse
|
6
|
Paiardi G, Richter S, Oreste P, Urbinati C, Rusnati M, Wade RC. The binding of heparin to spike glycoprotein inhibits SARS-CoV-2 infection by three mechanisms. J Biol Chem 2021; 298:101507. [PMID: 34929169 PMCID: PMC8683219 DOI: 10.1016/j.jbc.2021.101507] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 01/03/2023] Open
Abstract
Heparin, a naturally occurring glycosaminoglycan, has been found to have antiviral activity against SARS-CoV-2, the causative virus of COVID-19. To elucidate the mechanistic basis for the antiviral activity of heparin, we investigated the binding of heparin to the SARS-CoV-2 spike glycoprotein by means of sliding window docking, molecular dynamics simulations, and biochemical assays. Our simulations show that heparin binds at long, positively-charged patches on the spike glycoprotein, thereby masking basic residues of both the receptor binding domain (RBD) and the multifunctional S1/S2 site. Biochemical experiments corroborated the simulation results, showing that heparin inhibits the furin-mediated cleavage of spike by binding to the S1/S2 site. Our simulations also showed that heparin can act on the hinge region responsible for motion of the RBD between the inactive closed and active open conformations of the spike glycoprotein. In simulations of the closed spike homotrimer, heparin binds the RBD and the N-terminal domain of two adjacent spike subunits and hinders opening. In simulations of open spike conformations, heparin induces stabilization of the hinge region and a change in RBD motion. Taken together, our results indicate that heparin can inhibit SARS-CoV-2 infection by three mechanisms: by allosterically hindering binding to the host cell receptor, by directly competing with binding to host heparan sulfate proteoglycan co-receptors, and by preventing spike cleavage by furin. Furthermore, these simulations provide insights into how host heparan sulfate proteoglycans can facilitate viral infection. Our results will aid the rational optimization of heparin derivatives for SARS-CoV-2 antiviral therapy.
Collapse
Affiliation(s)
- Giulia Paiardi
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), 69118 Heidelberg, Germany; Macromolecular Interaction Analysis Unit, Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, 25123 Brescia, Italy.
| | - Stefan Richter
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), 69118 Heidelberg, Germany
| | | | - Chiara Urbinati
- Macromolecular Interaction Analysis Unit, Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, 25123 Brescia, Italy
| | - Marco Rusnati
- Macromolecular Interaction Analysis Unit, Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, 25123 Brescia, Italy
| | - Rebecca C Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), 69118 Heidelberg, Germany; Zentrum für Molekulare Biologie (ZMBH), DKFZ-ZMBH Alliance and Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, 69120 Heidelberg, Germany.
| |
Collapse
|
7
|
Chhabra M, Doherty GG, See NW, Gandhi NS, Ferro V. From Cancer to COVID-19: A Perspective on Targeting Heparan Sulfate-Protein Interactions. CHEM REC 2021; 21:3087-3101. [PMID: 34145723 PMCID: PMC8441866 DOI: 10.1002/tcr.202100125] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/01/2021] [Indexed: 12/16/2022]
Abstract
Heparan sulfate (HS) is a complex, polyanionic polysaccharide ubiquitously expressed on cell surfaces and in the extracellular matrix. HS interacts with numerous proteins to mediate a vast array of biological and pathological processes. Inhibition of HS-protein interactions is thus an attractive approach for new therapeutic development for cancer and infectious diseases, including COVID-19; however, synthesis of well-defined native HS oligosaccharides remains challenging. This has aroused significant interest in the development of HS mimetics which are more synthetically tractable and have fewer side effects, such as undesired anticoagulant activity. This account provides a perspective on the design and synthesis of different classes of HS mimetics with useful properties, and the development of various assays and molecular modelling tools to progress our understanding of their interactions with HS-binding proteins.
Collapse
Affiliation(s)
- Mohit Chhabra
- School of Chemistry and Molecular BiosciencesThe University of Queensland4072BrisbaneQLDAustralia
| | - Gareth G. Doherty
- School of Chemistry and Molecular BiosciencesThe University of Queensland4072BrisbaneQLDAustralia
| | - Nicholas W. See
- School of Chemistry and Molecular BiosciencesThe University of Queensland4072BrisbaneQLDAustralia
| | - Neha S. Gandhi
- School of Chemistry and PhysicsQueensland University of Technology4000BrisbaneQLDAustralia
| | - Vito Ferro
- School of Chemistry and Molecular BiosciencesThe University of Queensland4072BrisbaneQLDAustralia
| |
Collapse
|
8
|
Wang P, Zhao J, Hossaini Nasr S, Otieno SA, Zhang F, Qiang W, Linhardt RJ, Huang X. Probing Amyloid β Interactions with Synthetic Heparan Sulfate Oligosaccharides. ACS Chem Biol 2021; 16:1894-1899. [PMID: 33592143 DOI: 10.1021/acschembio.0c00904] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Heparan sulfate (HS) can play important roles in the biology and pathology of amyloid β (Aβ), a hallmark of Alzheimer's disease. To better understand the structure-activity relationship of HS/Aβ interactions, synthetic HS oligosaccharides ranging from tetrasaccharides to decasaccharides have been utilized to study Aβ interactions. Surface plasmon resonance experiments showed that the highly sulfated HS tetrasaccharides bearing full 2-O, 6-O, and N-sulfations exhibited the strongest binding with Aβ among the tetrasaccharides investigated. Elongating the glycan length to hexa- and deca-saccharides significantly enhanced Aβ affinity compared to the corresponding HS tetrasaccharide. Solid state NMR studies of the complexes of Aβ with HS hexa- and deca-saccharides showed most significant chemical shift perturbation in the C-terminus residues of Aβ. The strong binding HS oligosaccharides could reduce the cellular toxicities induced by Aβ. This study provides new insights into HS/Aβ interactions, highlighting how synthetic structurally well-defined HS oligosaccharides can assist in biological understanding of Aβ.
Collapse
Affiliation(s)
| | - Jing Zhao
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | | | - Sarah A. Otieno
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Fuming Zhang
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Wei Qiang
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Robert J. Linhardt
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | | |
Collapse
|
9
|
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
|
10
|
Systemically Administered Homing Peptide Targets Dystrophic Lesions and Delivers Transforming Growth Factor-β (TGFβ) Inhibitor to Attenuate Murine Muscular Dystrophy Pathology. Pharmaceutics 2021; 13:pharmaceutics13091506. [PMID: 34575582 PMCID: PMC8471674 DOI: 10.3390/pharmaceutics13091506] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/07/2021] [Accepted: 09/13/2021] [Indexed: 01/14/2023] Open
Abstract
Muscular dystrophy is a progressively worsening and lethal disease, where accumulation of functionality-impairing fibrosis plays a key pathogenic role. Transforming growth factor-β1 (TGFβ1) is a central signaling molecule in the development of fibrosis in muscular dystrophic humans and mice. Inhibition of TGFβ1 has proven beneficial in mouse models of muscular dystrophy, but the global strategies of TGFβ1 inhibition produce significant detrimental side effects. Here, we investigated whether murine muscular dystrophy lesion-specific inhibition of TGFβ1 signaling by the targeted delivery of therapeutic decorin (a natural TGFβ inhibitor) by a vascular homing peptide CAR (CARSKNKDC) would reduce skeletal muscle fibrosis and pathology and increase functional characteristics of skeletal muscle. We demonstrate that CAR peptide homes to dystrophic lesions with specificity in two muscular dystrophy models. Recombinant fusion protein consisting of CAR peptide and decorin homes selectively to sites of skeletal muscle damage in mdxDBA2/J and gamma-sarcoglycan deficient DBA2/J mice. This targeted delivery reduced TGFβ1 signaling as demonstrated by reduced nuclear pSMAD staining. Three weeks of targeted decorin treatment decreased both membrane permeability and fibrosis and improved skeletal muscle function in comparison to control treatments in the mdxD2 mice. These results show that selective delivery of decorin to the sites of skeletal muscle damage attenuates the progression of murine muscular dystrophy.
Collapse
|
11
|
Weant J, Eveleth DD, Subramaniam A, Jenkins-Eveleth J, Blaber M, Li L, Ornitz DM, Alimardanov A, Broadt T, Dong H, Vyas V, Yang X, Bradshaw RA. Regenerative responses of rabbit corneal endothelial cells to stimulation by fibroblast growth factor 1 (FGF1) derivatives, TTHX1001 and TTHX1114. Growth Factors 2021; 39:14-27. [PMID: 34879776 DOI: 10.1080/08977194.2021.2012468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Utilising rabbit corneal endothelial cells (CEC) in three different paradigms, two human FGF1 derivatives (TTHX1001 and TTHX1114), engineered to exhibit greater stability, were tested as proliferative agents. Primary CECs and mouse NIH 3T3 cells treated with the two FGF1 derivatives showed equivalent EC50 ranges (3.3-24 vs.1.9-16. ng/mL) and, in organ culture, chemically lesioned corneas regained half of the lost endothelial layer in three days after treatment with the FGF1 derivatives as compared to controls. In vivo, following cryolesioning, the CEC monolayer, as judged by specular microscopy, regenerated 10-11 days faster when treated with TTHX1001. Over two weeks, all treated eyes showed clearing of opacity about twice that of untreated controls. In all three rabbit models, both FGF1 derivatives were effective in inducing CEC proliferation over control conditions, supporting the prediction that these stabilised FGF1 derivatives can potentially regenerate corneal endothelial deficits in humans.
Collapse
Affiliation(s)
| | | | | | | | - Michael Blaber
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, USA
| | - Ling Li
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Asaf Alimardanov
- Therapeutics Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Trevor Broadt
- Biopharmaceutical Development Program, Advanced Technology Research Facility, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research Inc, Frederick, MD, USA
| | - Hui Dong
- Biopharmaceutical Development Program, Advanced Technology Research Facility, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research Inc, Frederick, MD, USA
| | - Vinay Vyas
- Biopharmaceutical Development Program, Advanced Technology Research Facility, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research Inc, Frederick, MD, USA
| | - Xiaoyi Yang
- Biopharmaceutical Development Program, Advanced Technology Research Facility, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research Inc, Frederick, MD, USA
| | - Ralph A Bradshaw
- Trefoil Therapeutics, Inc, San Diego, CA, USA
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
12
|
Chhabra M, Ferro V. PI-88 and Related Heparan Sulfate Mimetics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:473-491. [PMID: 32274723 DOI: 10.1007/978-3-030-34521-1_19] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The heparan sulfate mimetic PI-88 (muparfostat) is a complex mixture of sulfated oligosaccharides that was identified in the late 1990s as a potent inhibitor of heparanase. In preclinical animal models it was shown to block angiogenesis, metastasis and tumor growth, and subsequently became the first heparanase inhibitor to enter clinical trials for cancer. It progressed to Phase III trials but ultimately was not approved for use. Herein we summarize the preparation, physicochemical and biological properties of PI-88, and discuss preclinical/clinical and structure-activity relationship studies. In addition, we discuss the PI-88-inspired development of related HS mimetic heparanase inhibitors with improved properties, ultimately leading to the discovery of PG545 (pixatimod) which is currently in clinical trials.
Collapse
Affiliation(s)
- Mohit Chhabra
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| | - Vito Ferro
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia. .,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
13
|
Zhu S, Li J, Loka RS, Song Z, Vlodavsky I, Zhang K, Nguyen HM. Modulating Heparanase Activity: Tuning Sulfation Pattern and Glycosidic Linkage of Oligosaccharides. J Med Chem 2020; 63:4227-4255. [PMID: 32216347 DOI: 10.1021/acs.jmedchem.0c00156] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Heparanase cleaves polymeric heparan sulfate (HS) molecules into smaller oligosaccharides, allowing for release of angiogenic growth factors promoting tumor development and autoreactive immune cells to reach the insulin-producing β cells. Interaction of heparanase with HS chains is regulated by specific substrate sulfation sequences. We have synthesized 11 trisaccharides that are highly tunable in structure and sulfation pattern, allowing us to determine how heparanase recognizes HS substrate and selects a favorable cleavage site. Our study shows that (1) N-SO3- at +1 subsite and 6-O-SO3- at -2 subsite of trisaccharides are critical for heparanase recognition, (2) addition of 2-O-SO3- at the -1 subsite and of 3-O-SO3- to GlcN unit is not advantageous, and (3) the anomeric configuration (α or β) at the reducing end is crucial in controlling heparanase activity. Our study also illustrates that the α-trisaccharide having N- and 6-O-SO3- at -2 and +1 subsites inhibited heparanase and was resistant toward hydrolysis.
Collapse
Affiliation(s)
- Sanyong Zhu
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Jiayi Li
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Ravi S Loka
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Zhenfeng Song
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Hien M Nguyen
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| |
Collapse
|
14
|
Crijns H, Vanheule V, Proost P. Targeting Chemokine-Glycosaminoglycan Interactions to Inhibit Inflammation. Front Immunol 2020; 11:483. [PMID: 32296423 PMCID: PMC7138053 DOI: 10.3389/fimmu.2020.00483] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
Leukocyte migration into tissues depends on the activity of chemokines that form concentration gradients to guide leukocytes to a specific site. Interaction of chemokines with their specific G protein-coupled receptors (GPCRs) on leukocytes induces leukocyte adhesion to the endothelial cells, followed by extravasation of the leukocytes and subsequent directed migration along the chemotactic gradient. Interaction of chemokines with glycosaminoglycans (GAGs) is crucial for extravasation in vivo. Chemokines need to interact with GAGs on endothelial cells and in the extracellular matrix in tissues in order to be presented on the endothelium of blood vessels and to create a concentration gradient. Local chemokine retention establishes a chemokine gradient and prevents diffusion and degradation. During the last two decades, research aiming at reducing chemokine activity mainly focused on the identification of inhibitors of the interaction between chemokines and their cognate GPCRs. This approach only resulted in limited success. However, an alternative strategy, targeting chemokine-GAG interactions, may be a promising approach to inhibit chemokine activity and inflammation. On this line, proteins derived from viruses and parasites that bind chemokines or GAGs may have the potential to interfere with chemokine-GAG interactions. Alternatively, chemokine mimetics, including truncated chemokines and mutant chemokines, can compete with chemokines for binding to GAGs. Such truncated or mutated chemokines are characterized by a strong binding affinity for GAGs and abrogated binding to their chemokine receptors. Finally, Spiegelmers that mask the GAG-binding site on chemokines, thereby preventing chemokine-GAG interactions, were developed. In this review, the importance of GAGs for chemokine activity in vivo and strategies that could be employed to target chemokine-GAG interactions will be discussed in the context of inflammation.
Collapse
Affiliation(s)
- Helena Crijns
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Vincent Vanheule
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
|
16
|
Loka RS, Sletten ET, Barash U, Vlodavsky I, Nguyen HM. Specific Inhibition of Heparanase by a Glycopolymer with Well-Defined Sulfation Pattern Prevents Breast Cancer Metastasis in Mice. ACS APPLIED MATERIALS & INTERFACES 2019; 11:244-254. [PMID: 30543095 PMCID: PMC6512314 DOI: 10.1021/acsami.8b17625] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Heparanase, the heparan sulfate polysaccharide degrading endoglycosidase enzyme, has been correlated with tumor angiogenesis and metastasis and therefore has become a potential target for anticancer drug development. In this systematic study, the sulfation pattern of the pendant disaccharide moiety on synthetic glycopolymers was synthetically manipulated to achieve optimal heparanase inhibition. Upon evaluation, a glycopolymer with 12 repeating units was determined to be the most potent inhibitor of heparanase (IC50 = 0.10 ± 0.36 nM). This glycopolymer was further examined for cross-bioactivity using a solution-based competitive biolayer interferometry assay with other HS-binding proteins (growth factors, P-selectin, and platelet factor 4), which are responsible for mediating angiogenic activity, cell metastasis, and antibody-induced thrombocytopenia. The synthetic glycopolymer has low affinity for these HS-binding proteins in comparison to natural heparin. In addition, the glycopolymer possessed no proliferative properties toward human umbilical endothelial cells (HUVECs) and a potent antimetastatic effect against 4T1 mammary carcinoma cells. Thus, our study not only establishes a specific inhibitor of heparanase with high affinity but also illustrates the high effectiveness of this multivalent heparanase inhibitor in inhibiting experimental metastasis in vivo.
Collapse
Affiliation(s)
- Ravi S Loka
- Department of Chemistry , Wayne State University , Detroit , Michigan 48202 , United States
| | - Eric T Sletten
- Department of Chemistry , University of Iowa , Iowa City, Iowa 52242 , United States
| | - Uri Barash
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine , Technion-Israel Institute of Technology , Haifa , Israel
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine , Technion-Israel Institute of Technology , Haifa , Israel
| | - Hien M Nguyen
- Department of Chemistry , Wayne State University , Detroit , Michigan 48202 , United States
| |
Collapse
|
17
|
Prado E, Bonnat L, Bonnet H, Lavergne T, Van der Heyden A, Pratviel G, Dejeu J, Defrancq E. Influence of the SPR Experimental Conditions on the G-Quadruplex DNA Recognition by Porphyrin Derivatives. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:13057-13064. [PMID: 30293430 DOI: 10.1021/acs.langmuir.8b02942] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Surface plasmon resonance (SPR) is a powerful technique to study the interactions of ligands with analytes and therefore a number of biosensor surfaces and injection methods have been developed so far. However, many experimental parameters can affect the interactions and consequently the affinity measurements. In particular, the interactions of positively charged analytes (often used for anionic nucleic acids targets) can be influenced by the sensing surfaces (e.g., negatively charged), leading to significant nonspecific interactions as well as regeneration problems. The aim of the present work is to investigate the effect of different parameters, including ionic strength, SPR biosensor (i.e., nature of the surfaces), and the injection method on the recognition of porphyrin G-quadruplex ligands. We demonstrate that the injection method does not influence the affinity whereas the ionic strength and the nature of the surface impact the recognition properties of the porphyrin for the G-quadruplex DNA. We also found that self-assembled monolayer coating surface presents many advantages in comparison with carboxymethylated dextran surface for SPR studies of G-quadruplex DNA/ligand interactions: (i) the electrostatic interaction with charged analytes is less important, (ii) its structure/composition is less sensitive to the ionic concentration and less prone to unspecific adsorption, (iii) it is easily homemade, and (iv) the cost is approximately 10 times cheaper.
Collapse
Affiliation(s)
- E Prado
- Univ. Grenoble Alpes, CNRS, DCM , 38000 Grenoble , France
| | - L Bonnat
- Univ. Grenoble Alpes, CNRS, DCM , 38000 Grenoble , France
| | - H Bonnet
- Univ. Grenoble Alpes, CNRS, DCM , 38000 Grenoble , France
| | - T Lavergne
- Univ. Grenoble Alpes, CNRS, DCM , 38000 Grenoble , France
| | | | - G Pratviel
- CNRS, Laboratoire de Chimie de Coordination , 205 route de Narbonne, BP44099 , F-31077 Toulouse Cedex 4, France
- Université de Toulouse, UPS, INPT , Toulouse , France
| | - J Dejeu
- Univ. Grenoble Alpes, CNRS, DCM , 38000 Grenoble , France
| | - E Defrancq
- Univ. Grenoble Alpes, CNRS, DCM , 38000 Grenoble , France
| |
Collapse
|
18
|
Lin L, Xu L, Xiao C, Zhou L, Gao N, Wu M, Zhao J. Plasma contact activation by a fucosylated chondroitin sulfate and its structure–activity relationship study. Glycobiology 2018; 28:754-764. [PMID: 30016441 DOI: 10.1093/glycob/cwy067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 07/13/2018] [Indexed: 12/26/2022] Open
Affiliation(s)
- Lisha Lin
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, College of Life Sciences, Beijing, China
| | - Li Xu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, College of Life Sciences, Beijing, China
| | - Chuang Xiao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, College of Life Sciences, Beijing, China
| | - Lutan Zhou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, College of Life Sciences, Beijing, China
| | - Na Gao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Mingyi Wu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Jinhua Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
19
|
Modulating the degree of fucosylation of fucosylated chondroitin sulfate enhances heparin cofactor II-dependent thrombin inhibition. Eur J Med Chem 2018; 154:133-143. [PMID: 29787913 DOI: 10.1016/j.ejmech.2018.05.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/30/2018] [Accepted: 05/15/2018] [Indexed: 12/24/2022]
Abstract
Fucosylated chondroitin sulfate (FCS), an unusual glycosaminoglycan with fucose side chains, is a promising anticoagulant agent. To assess the effect of its structure on anticoagulant activity, its derivatives with various degrees of fucosylation (DF), molecular weights (Mw) and sulfation patterns were prepared and characterized. Biological tests showed that their APTT (activated partial thromboplastin time) prolonging activity and intrinsic factor Xase complex (factor IXa-VIIIa-Ca2+-PL complex) inhibitory activity were both reduced in FCS derivatives with lower Mw and DF. However, FCSs with DF at least 16% resulted in greater heparin cofactor II (HCII)-dependent thrombin inhibitory activity in response to decreasing DF, and these activities did not depend on Mw (Mw > 5.2 kDa). Solution competition binding assay further suggested that modulating the DF of FCS derivatives might enhance inhibition of thrombin by activating HCII. These findings imply that FCS derivatives with suitable chain length and DF value may be novel anticoagulants by activating HCII.
Collapse
|
20
|
Katner SJ, Johnson WE, Peterson EJ, Page P, Farrell NP. Comparison of Metal-Ammine Compounds Binding to DNA and Heparin. Glycans as Ligands in Bioinorganic Chemistry. Inorg Chem 2018; 57:3116-3125. [PMID: 29473748 DOI: 10.1021/acs.inorgchem.7b03043] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We present spectroscopic and biophysical approaches to examine the affinity of metal-ammine coordination complexes for heparin as a model for heparan sulfate (HS). Similar to nucleic acids, the highly anionic nature of heparin means it is associated in vivo with physiologically relevant cations, and this work extends their bioinorganic chemistry to substitution-inert metal-ammine compounds (M). Both indirect and direct assays were developed. M compounds are competitive inhibitors of methylene blue (MB)-heparin binding, and the change in the absorbance of the dye in the presence or absence of heparin can be used as an indirect reporter of M-heparin affinity. A second indirect assay uses the change in fluorescence of TAMRA-R9, a nonaarginine linked to a fluorescent TAMRA moiety, as a reporter for M-heparin binding. Direct assays are surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC). The Kd values for TriplatinNC-heparin varied to some extent depending on the technique from 33.1 ± 2 nM (ITC) to 66.4 ± 1.3 nM (MB absorbance assay) and 340 ± 30 nM (SPR). The differences are explained by the nature of the technique and the use of heparin of differing molecular weight. Indirect probes using the displacement of ethidium bromide from DNA or, separately, fluorescently labeled oligonucleotide (DNA-Fl) can measure the relative affinities of heparin and DNA for M compounds. These assays showed essentially equivalent affinity of TriplatinNC for heparin and DNA. The generality of these methods was confirmed with a series of mononuclear cobalt, ruthenium, and platinum compounds with significantly lower affinity because of their smaller overall positive charge but in the order [Co(NH3)6]3+ > [Ru(NH3)6]3+ > [Pt(NH3)4]2+. The results on heparin can be extrapolated to glycosoaminoglycans such as HS, emphasizing the relevance of glycan interactions in understanding the biological properties of coordination compounds and the utility of the metalloglycomics concept for extending bioinorganic chemistry to this class of important biomolecules.
Collapse
Affiliation(s)
- Samantha J Katner
- Department of Chemistry and Massey Cancer Center , Virginia Commonwealth University (VCU) , Richmond , Virginia 23284 , United States
| | - Wyatt E Johnson
- Department of Chemistry and Massey Cancer Center , Virginia Commonwealth University (VCU) , Richmond , Virginia 23284 , United States
| | - Erica J Peterson
- Department of Chemistry and Massey Cancer Center , Virginia Commonwealth University (VCU) , Richmond , Virginia 23284 , United States
| | - Phillip Page
- Reichert Technologies , Depew , New York 14043 , United States
| | - Nicholas P Farrell
- Department of Chemistry and Massey Cancer Center , Virginia Commonwealth University (VCU) , Richmond , Virginia 23284 , United States
| |
Collapse
|
21
|
Interactions between depolymerized fucosylated glycosaminoglycan and coagulation proteases or inhibitors. Thromb Res 2016; 146:59-68. [PMID: 27611497 DOI: 10.1016/j.thromres.2016.08.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 08/12/2016] [Accepted: 08/26/2016] [Indexed: 01/24/2023]
Abstract
Fucosylated glycosaminoglycan (FG) is a structurally novel glycosaminoglycan derivative, and it has potent anticoagulant activity. Depolymerized FG (dFG) is a selective factor Xase (FXase, FIXa-FVIIIa complex) inhibitor and it has antithrombotic action without major bleeding risks. In this study, we report the effects of dFG-3 (Mw ~14kDa) on the catalysis rates of factor IIa (FIIa), factor Xa (FXa) and factor IXa (FIXa) inhibition by antithrombin (AT), and the kinetic of the interactions between coagulation proteases or inhibitors and dFG-3 were also studied using biolayer interferometry (BLI) technology. We found that dFG-3 had much weaker catalysis activity of coagulation proteases inhibition by AT compared with heparin (UFH). The binding affinity of AT bound to dFG-3 was lower than UFH, and the UFH-AT interaction fitted well with biphasic-binding model while dFG-3-AT interaction was monophasic-binding, suggesting dFG-3 might not have allosteric activation effect on AT. The results are consistent with AT-independent inhibitory activities of dFG-3. dFG-3 could strongly bind to FIXa with much higher affinity than UFH, further explained the reason for its potent FXase inhibitory activity. Additionally, the binding ability of dFG-3 and FIXa decreased with decreasing molecular, and the fucose side chains and carboxyl groups of dFG-3 might be required for its high affinity binding with FIXa. Our data supports further the investigation of dFG-3 as a promising anticoagulant drug inhibiting the intrinsic FXase by binding to FIXa.
Collapse
|
22
|
Kuhnast B, El Hadri A, Boisgard R, Hinnen F, Richard S, Caravano A, Nancy-Portebois V, Petitou M, Tavitian B, Dollé F. Synthesis, radiolabeling with fluorine-18 and preliminary in vivo evaluation of a heparan sulphate mimetic as potent angiogenesis and heparanase inhibitor for cancer applications. Org Biomol Chem 2016; 14:1915-20. [DOI: 10.1039/c5ob02513c] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A rationally designed, fully synthetic, octasaccharide-based, HS mimetic has been synthesized, in vitro characterized, labeled with fluorine-18, and in vivo imaged with PET in rats.
Collapse
Affiliation(s)
- B. Kuhnast
- CEA
- Institut d'imagerie biomédicale
- Service Hospitalier Frédéric Joliot
- 91400 Orsay
- France
| | - A. El Hadri
- Endotis Pharma
- Biocitech Park
- 93230 Romainville
- France
- CarboMimetics
| | - R. Boisgard
- CEA
- Institut d'imagerie biomédicale
- Service Hospitalier Frédéric Joliot
- 91400 Orsay
- France
| | - F. Hinnen
- CEA
- Institut d'imagerie biomédicale
- Service Hospitalier Frédéric Joliot
- 91400 Orsay
- France
| | - S. Richard
- Endotis Pharma
- Biocitech Park
- 93230 Romainville
- France
| | - A. Caravano
- Endotis Pharma
- Biocitech Park
- 93230 Romainville
- France
| | | | - M. Petitou
- Endotis Pharma
- Biocitech Park
- 93230 Romainville
- France
| | - B. Tavitian
- Laboratoire PARCC UMR 970 Inserm/Université Paris Descartes
- Sorbonne Paris Cité
- Assistance Publique – Hôpitaux de Paris
- Hôpital Européen Georges Pompidou
- 75015 Paris
| | - F. Dollé
- CEA
- Institut d'imagerie biomédicale
- Service Hospitalier Frédéric Joliot
- 91400 Orsay
- France
| |
Collapse
|
23
|
Heparin/Heparan sulfate proteoglycans glycomic interactome in angiogenesis: biological implications and therapeutical use. Molecules 2015; 20:6342-88. [PMID: 25867824 PMCID: PMC6272510 DOI: 10.3390/molecules20046342] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis, the process of formation of new blood vessel from pre-existing ones, is involved in various intertwined pathological processes including virus infection, inflammation and oncogenesis, making it a promising target for the development of novel strategies for various interventions. To induce angiogenesis, angiogenic growth factors (AGFs) must interact with pro-angiogenic receptors to induce proliferation, protease production and migration of endothelial cells (ECs). The action of AGFs is counteracted by antiangiogenic modulators whose main mechanism of action is to bind (thus sequestering or masking) AGFs or their receptors. Many sugars, either free or associated to proteins, are involved in these interactions, thus exerting a tight regulation of the neovascularization process. Heparin and heparan sulfate proteoglycans undoubtedly play a pivotal role in this context since they bind to almost all the known AGFs, to several pro-angiogenic receptors and even to angiogenic inhibitors, originating an intricate network of interaction, the so called "angiogenesis glycomic interactome". The decoding of the angiogenesis glycomic interactome, achievable by a systematic study of the interactions occurring among angiogenic modulators and sugars, may help to design novel antiangiogenic therapies with implications in the cure of angiogenesis-dependent diseases.
Collapse
|
24
|
Angiogenic growth factors interactome and drug discovery: The contribution of surface plasmon resonance. Cytokine Growth Factor Rev 2014; 26:293-310. [PMID: 25465594 DOI: 10.1016/j.cytogfr.2014.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 11/10/2014] [Accepted: 11/11/2014] [Indexed: 11/21/2022]
Abstract
Angiogenesis is implicated in several pathological conditions, including cancer, and in regenerative processes, including the formation of collateral blood vessels after stroke. Physiological angiogenesis is the outcome of a fine balance between the action of angiogenic growth factors (AGFs) and anti-angiogenic molecules, while pathological angiogenesis occurs when this balance is pushed toward AGFs. AGFs interact with multiple endothelial cell (EC) surface receptors inducing cell proliferation, migration and proteases upregulation. On the contrary, free or extracellular matrix-associated molecules inhibit angiogenesis by sequestering AGFs (thus hampering EC stimulation) or by interacting with specific EC receptors inducing apoptosis or decreasing responsiveness to AGFs. Thus, angiogenesis results from an intricate network of interactions among pro- and anti-angiogenic molecules, EC receptors and various modulators. All these interactions represent targets for the development of pro- or anti-angiogenic therapies. These aims call for suitable technologies to study the countless interactions occurring during neovascularization. Surface plasmon resonance (SPR) is a label-free optical technique to study biomolecular interactions in real time. It has become the golden standard technology for interaction analysis in biomedical research, including angiogenesis. From a survey of the literature it emerges that SPR has already contributed substantially to the better understanding of the neovascularization process, laying the basis for the decoding of the angiogenesis "interactome" and the identification of "hub molecules" that may represent preferential targets for an efficacious modulation of angiogenesis. Here, the still unexploited full potential of SPR is enlightened, pointing to improvements in its use for a deeper understanding of the mechanisms of neovascularization and the identification of novel anti-angiogenic drugs.
Collapse
|
25
|
Yip CW, Cheung PFY, Leung ICY, Wong NCL, Cheng CKC, Fan ST, Cheung ST. Granulin-epithelin precursor interacts with heparan sulfate on liver cancer cells. Carcinogenesis 2014; 35:2485-94. [PMID: 25115442 PMCID: PMC4216055 DOI: 10.1093/carcin/bgu164] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The first study to demonstrate HS to affect GEP binding on the cell surface. Granulin-epithelin precursor (GEP) is a pluripotent secretory growth factor which promotes cancer progression in a number of human cancers. However, how cancer cells interact with GEP remains unknown. In this study, we aimed to identify the cell surface-binding partner of GEP on liver cancer cells. Human recombinant GEP (rGEP) was expressed and purified to homogeneity. The rGEP was shown to trigger phosphorylation of AKT and ERK1/2 in liver cancer cells. We demonstrated cell surface attachment of rGEP, which was blocked by prebinding of platelet-derived growth factor-AA, platelet-derived growth factor-BB and fibroblast growth factor-2. Therefore, heparan sulfate (HS) had been reasoned as the binding partner of rGEP. Heparinase digestion validated the role of HS on supporting the attachment. The heparin-binding domain of GEP was mapped to RRH(555-557) in the C-terminal region. Suppression of the HS polymerase exostosin-1 reduced the rGEP binding and rGEP-mediated signaling transduction. Suppression of a specific HS proteoglycan, glypican-3, also showed a partial reduction of rGEP binding and an inhibition on rGEP-mediated activation of AKT. Furthermore, glypican-3 was shown to correlate with the expressions of GEP in clinical samples (Spearman’s ρ = 0.363, P = 0.001). This study identified HS, partly through glypican-3, as a novel binding partner of GEP on the surface of liver cancer cells.
Collapse
Affiliation(s)
- Chi Wai Yip
- Department of Surgery, Centre for Cancer Research and
| | | | | | | | | | | | | |
Collapse
|
26
|
Gesslbauer B, Theuer M, Schweiger D, Adage T, Kungl AJ. New targets for glycosaminoglycans and glycosaminoglycans as novel targets. Expert Rev Proteomics 2013; 10:77-95. [PMID: 23414361 DOI: 10.1586/epr.12.75] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Biological functions of a variety of proteins are mediated via their interaction with glycosaminoglycans (GAGs). The structural diversity within the wide GAG landscape provides individual interaction sites for a multitude of proteins involved in several pathophysiological processes. This 'GAG angle' of such proteins as well as their specific GAG ligands give rise to novel therapeutic concepts for drug development. Current glycomic technologies to elucidate the glycan structure-function relationships, methods to investigate the selectivity and specificity of glycan-protein interactions and existing therapeutic approaches to interfere with GAG-protein interactions are discussed.
Collapse
Affiliation(s)
- Bernd Gesslbauer
- ProtAffin Biotechnologie AG, Reininghausstrasse 13a, 8020 Graz, Austria
| | | | | | | | | |
Collapse
|
27
|
Day ES, Capili AD, Borysenko CW, Zafari M, Whitty A. Determining the affinity and stoichiometry of interactions between unmodified proteins in solution using Biacore. Anal Biochem 2013; 440:96-107. [PMID: 23711722 DOI: 10.1016/j.ab.2013.05.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 05/03/2013] [Accepted: 05/10/2013] [Indexed: 11/30/2022]
Abstract
We describe a general Biacore method for measuring equilibrium binding affinities and stoichiometries for interactions between unmodified proteins and their unmodified ligands free in solution. Mixtures of protein and ligand are preequilibrated at different ratios in solution and then analyzed by Biacore using a sensor chip surface that detects only unbound analyte. Performing the Biacore analysis under mass transport limited conditions allows the concentration of unbound analyte to be determined from the initial velocity of binding. Plots of initial velocity versus the concentration of the varied binding partner are fitted to a quadratic binding equation to give the affinity and stoichiometry of binding. We demonstrate the method using soluble Her2 extracellular domain binding to monovalent, bivalent, and trivalent forms of an anti-Her2 antibody. The affinity we measured agrees with that obtained from conventional Biacore kinetic analysis, and the stoichiometries for the resulting 1:1, 1:2, and 1:3 complexes were confirmed by gel filtration with in-line light scattering. The method is applicable over an affinity range of approximately 100 pM to 1 μM and is particularly useful when there is concern that covalently modifying one or the other binding partner might affect its binding properties or where multivalency might otherwise complicate a quantitative analysis of binding.
Collapse
|
28
|
Chen Y, Scully M, Dawson G, Goodwin C, Xia M, Lu X, Kakkar A. Perturbation of the heparin/heparin-sulfate interactome of human breast cancer cells modulates pro-tumourigenic effects associated with PI3K/Akt and MAPK/ERK signalling. Thromb Haemost 2013; 109:1148-57. [PMID: 23571852 DOI: 10.1160/th12-12-0935] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 02/27/2013] [Indexed: 12/18/2022]
Abstract
Heparansulfate-proteoglycans (HSPGs) interact via their polyanionic heparansulfate (HS) side chains with a variety of proteins on the cell surface or within the extracellular matrix membrane. The large number of heparin/HS binding proteins form a highly interconnected functional network, which has been termed as the heparin/HS interactome and is functionally linked to physiological and pathological processes. The aim of this study was to investigate the global effect of these protein-HSPG interactions on the tumourigenicity of two breast cancer cell lines (MCF-7 and MDA-MB-231). Cancer cells were cultured in serum-free medium and treated with a concentration of heparin which was capable of modulating HS/ligand interaction. Microarray analysis of MCF-7 cells cultured under these conditions showed that expression of 105 of 1,357 genes potentially related to the pathogenesis of breast neoplasm was significantly altered by heparin treatment. The changes in gene expression correlated with a less tumourigenic phenotype, including reduction of cell adhesive, invasive and migratory properties. These effects were associated with an inhibition of the PI3K/Akt and Raf/MEK/ERK signalling pathways. The modulatory effect of heparin on HS-associated activity was confirmed with one example of heparin/HS interactomes, transforming growth factor β (TGFβ). The innate TGFβ activity of MCF-7 cells was reduced by heparin treatment, with specific interruption of the TGFβ-Smad signalling pathway. The pro-tumourigenic contribution of the heparin/HS interactomes was verified in cells in which HSPG synthesis was blocked using β-xyloside. In conclusion, the interaction between cell surface HPSGs and innate heparin/HS interactomes makes a significant contribution to the tumourigenicity.
Collapse
Affiliation(s)
- Yunliang Chen
- Thrombosis Research Institute, Emmanuel Kaye Building, Manresa Road, London SW3 6LR, UK
| | | | | | | | | | | | | |
Collapse
|
29
|
Liu L, Li C, Cochran S, Feder D, Guddat LW, Ferro V. A focused sulfated glycoconjugate Ugi library for probing heparan sulfate-binding angiogenic growth factors. Bioorg Med Chem Lett 2012; 22:6190-4. [DOI: 10.1016/j.bmcl.2012.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 07/23/2012] [Accepted: 08/01/2012] [Indexed: 11/30/2022]
|
30
|
Liu L, Li C, Cochran S, Jimmink S, Ferro V. Synthesis of a Heparan Sulfate Mimetic Library Targeting FGF and VEGF via Click Chemistry on a Monosaccharide Template. ChemMedChem 2012; 7:1267-75. [DOI: 10.1002/cmdc.201200151] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 04/26/2012] [Indexed: 11/07/2022]
|
31
|
Ferro V, Liu L, Johnstone KD, Wimmer N, Karoli T, Handley P, Rowley J, Dredge K, Li CP, Hammond E, Davis K, Sarimaa L, Harenberg J, Bytheway I. Discovery of PG545: A Highly Potent and Simultaneous Inhibitor of Angiogenesis, Tumor Growth, and Metastasis. J Med Chem 2012; 55:3804-13. [DOI: 10.1021/jm201708h] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Vito Ferro
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| | - Ligong Liu
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| | - Ken D. Johnstone
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| | - Norbert Wimmer
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| | - Tomislav Karoli
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| | - Paul Handley
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| | - Jessica Rowley
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| | - Keith Dredge
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| | - Cai Ping Li
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| | - Edward Hammond
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| | - Kat Davis
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| | - Laura Sarimaa
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| | - Job Harenberg
- Clinical
Pharmacology, Faculty
of Medicine Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Ian Bytheway
- Drug Design Group, Progen Pharmaceuticals Limited, Brisbane, Queensland
4076, Australia
| |
Collapse
|
32
|
Rich RL, Myszka DG. Survey of the 2009 commercial optical biosensor literature. J Mol Recognit 2012; 24:892-914. [PMID: 22038797 DOI: 10.1002/jmr.1138] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We took a different approach to reviewing the commercial biosensor literature this year by inviting 22 biosensor users to serve as a review committee. They set the criteria for what to expect in a publication and ultimately decided to use a pass/fail system for selecting which papers to include in this year's reference list. Of the 1514 publications in 2009 that reported using commercially available optical biosensor technology, only 20% passed their cutoff. The most common criticism the reviewers had with the literature was that "the biosensor experiments could have been done better." They selected 10 papers to highlight good experimental technique, data presentation, and unique applications of the technology. This communal review process was educational for everyone involved and one we will not soon forget.
Collapse
Affiliation(s)
- Rebecca L Rich
- Center for Biomolecular Interaction Analysis, University of Utah, Salt Lake City, UT, USA
| | | |
Collapse
|
33
|
Zhao W, McCallum SA, Xiao Z, Zhang F, Linhardt RJ. Binding affinities of vascular endothelial growth factor (VEGF) for heparin-derived oligosaccharides. Biosci Rep 2012; 32:71-81. [PMID: 21658003 PMCID: PMC3190640 DOI: 10.1042/bsr20110077] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Heparin and HS (heparan sulfate) exert their wide range of biological activities by interacting with extracellular protein ligands. Among these important protein ligands are various angiogenic growth factors and cytokines. HS binding to VEGF (vascular endothelial growth factor) regulates multiple aspects of vascular development and function through its specific interaction with HS. Many studies have focused on HS-derived or HS-mimicking structures for the characterization of VEGF165 interaction with HS. Using a heparinase 1-prepared small library of heparin-derived oligosaccharides ranging from hexasaccharide to octadecasaccharide, we systematically investigated the heparin-specific structural features required for VEGF binding. We report the apparent affinities for the association between the heparin-derived oligosaccharides with both VEGF165 and VEGF55, a peptide construct encompassing exclusively the heparin-binding domain of VEGF165. An octasaccharide was the minimum size of oligosaccharide within the library to efficiently bind to both forms of VEGF and a tetradecasaccharide displayed an effective binding affinity to VEGF165 comparable to unfractionated heparin. The range of relative apparent binding affinities among VEGF and the panel of heparin-derived oligosaccharides demonstrate that the VEGF binding affinity likely depends on the specific structural features of these oligosaccharides, including their degree of sulfation, sugar-ring stereochemistry and conformation. Notably, the unique 3-O-sulfo group found within the specific antithrombin binding site of heparin is not required for VEGF165 binding. These findings afford new insight into the inherent kinetics and affinities for VEGF association with heparin and heparin-derived oligosaccharides with key residue-specific modifications and may potentially benefit the future design of oligosaccharide-based anti-angiogenesis drugs.
Collapse
Affiliation(s)
- Wenjing Zhao
- Department of Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Scott A. McCallum
- Department of Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Zhongping Xiao
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Fuming Zhang
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Robert J. Linhardt
- Department of Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| |
Collapse
|
34
|
Desai BJ, Boothello RS, Mehta AY, Scarsdale JN, Wright HT, Desai UR. Interaction of thrombin with sucrose octasulfate. Biochemistry 2011; 50:6973-82. [PMID: 21736375 PMCID: PMC3172994 DOI: 10.1021/bi2004526] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The serine protease thrombin plays multiple roles in many important physiological processes, especially coagulation, where it functions as both a pro- and anticoagulant. The polyanionic glycosaminoglycan heparin modulates thrombin's activity through binding at exosite II. Sucrose octasulfate (SOS) is often used as a surrogate for heparin, but it is not known whether it is an effective heparin mimic in its interaction with thrombin. We have characterized the interaction of SOS with thrombin in solution and determined a crystal structure of their complex. SOS binds thrombin with a K(d) of ~1.4 μM, comparable to that of the much larger polymeric heparin measured under the same conditions. Nonionic (hydrogen bonding) interactions make a larger contribution to thrombin binding of SOS than to heparin. SOS binding to exosite II inhibits thrombin's catalytic activity with high potency but with low efficacy. Analytical ultracentrifugation shows that bovine and human thrombins are monomers in solution in the presence of SOS, in contrast to their complexes with heparin, which are dimers. In the X-ray crystal structure, two molecules of SOS are bound nonequivalently to exosite II portions of a thrombin dimer, in contrast to the 1:2 stoichiometry of the heparin-thrombin complex, which has a different monomer association mode in the dimer. SOS and heparin binding to exosite II of thrombin differ on both chemical and structural levels and, perhaps most significantly, in thrombin inhibition. These differences may offer paths to the design of more potent exosite II binding, allosteric small molecules as modulators of thrombin function.
Collapse
Affiliation(s)
- Bijoy J. Desai
- Departments of Medicinal Chemistry, Biochemistry and Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University, 800 E. Leigh Street, Suite 212, Richmond, VA 23219
| | - Rio S. Boothello
- Departments of Medicinal Chemistry, Biochemistry and Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University, 800 E. Leigh Street, Suite 212, Richmond, VA 23219
| | - Akul Y. Mehta
- Departments of Medicinal Chemistry, Biochemistry and Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University, 800 E. Leigh Street, Suite 212, Richmond, VA 23219
| | - J. Neel Scarsdale
- Departments of Medicinal Chemistry, Biochemistry and Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University, 800 E. Leigh Street, Suite 212, Richmond, VA 23219
| | - H. Tonie Wright
- Departments of Medicinal Chemistry, Biochemistry and Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University, 800 E. Leigh Street, Suite 212, Richmond, VA 23219
| | - Umesh R. Desai
- Departments of Medicinal Chemistry, Biochemistry and Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University, 800 E. Leigh Street, Suite 212, Richmond, VA 23219
| |
Collapse
|
35
|
Anderson SM, Shergill B, Barry ZT, Manousiouthakis E, Chen TT, Botvinick E, Platt MO, Iruela-Arispe ML, Segura T. VEGF internalization is not required for VEGFR-2 phosphorylation in bioengineered surfaces with covalently linked VEGF. Integr Biol (Camb) 2011; 3:887-96. [PMID: 21826315 DOI: 10.1039/c1ib00037c] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Vascular endothelial growth factor (VEGF) is known to activate proliferation, migration, and survival pathways in endothelial cells through phosphorylation of VEGF receptor-2 (VEGFR-2). VEGF has been incorporated into biomaterials through encapsulation, electrostatic sequestration, and covalent attachment, but the effect of these immobilization strategies on VEGF signaling has not been thoroughly investigated. Further, although growth factor internalization along with the receptor generally occurs in a physiological setting, whether this internalization is needed for receptor phosphorylation is not entirely clear. Here we show that VEGF covalently bound through a modified heparin molecule elicits an extended response of pVEGFR-2 in human umbilical vein endothelial cells (HUVECs) and that the covalent linkage reduces internalization of the growth factor during receptor endocytosis. Optical tweezer measurements show that the rupture force required to disrupt the heparin-VEGF-VEGFR-2 interaction increases from 3-8 pN to 6-12 pN when a covalent bond is introduced between VEGF and heparin. Importantly, by covalently binding VEGF to a heparin substrate, the stability (half-life) of VEGF is extended over three-fold. Here, mathematical models support the biological conclusions, further suggesting that VEGF internalization is significantly reduced when covalently bound, and indicating that VEGF is available for repeated phosphorylation events.
Collapse
Affiliation(s)
- Sean M Anderson
- Department of Chemical and Biomolecular Engineering, University of California, 420 Westwood Plaza, 5531 Boelter Hall, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Collier JH, Segura T. Evolving the use of peptides as components of biomaterials. Biomaterials 2011; 32:4198-204. [PMID: 21515167 PMCID: PMC3389831 DOI: 10.1016/j.biomaterials.2011.02.030] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 02/12/2011] [Indexed: 01/05/2023]
Abstract
This manuscript is part of a debate on the statement that "the use of short synthetic adhesion peptides, like RGD, is the best approach in the design of biomaterials that guide cell behavior for regenerative medicine and tissue engineering". We take the position that although there are some acknowledged disadvantages of using short peptide ligands within biomaterials, it is not necessary to discard the notion of using peptides within biomaterials entirely, but rather to reinvent and evolve their use. Peptides possess advantageous chemical definition, access to non-native chemistries, amenability to de novo design, and applicability within parallel approaches. Biomaterials development programs that require such aspects may benefit from a peptide-based strategy.
Collapse
Affiliation(s)
- Joel H. Collier
- Assistant Professor, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Mail Code 5032, Chicago, IL 60637, (773) 834-4161, (773) 834-4546 (fax)
| | - Tatiana Segura
- Assistant Professor, 420 Westwood Plaza, 5531 Boelter Hall, Los Angeles, CA 90095, (310) 206 3980
| |
Collapse
|
37
|
Abstract
Abstract
CXCL4 and CXCL4L1 are 2 closely related CXC chemokines that exhibit potent antiangiogenic activity. Because interactions with glycosaminoglycans play a crucial role in chemokines activity, we determined the binding parameters of CXCL4 and CXCL4L1 for heparin, heparan sulfate, and chondroitin sulfate B. We further demonstrated that the Leu67/His67 substitution is critical for the decrease in glycan binding of CXCL4L1 but also for the increase of its angiostatic activities. Using a set of mutants, we show that glycan affinity and angiostatic properties are not completely related. These data are reinforced using a monoclonal antibody that specifically recognizes structural modifications in CXCL4L1 due to the presence of His67 and that blocks its biologic activity. In vivo, half-life and diffusibility of CXCL4L1 compared with CXCL4 is strongly increased. As opposed to CXCL4L1, CXCL4 is preferentially retained at its site of expression. These findings establish that, despite small differences in the primary structure, CXCL4L1 is highly distinct from CXCL4. These observations are not only of great significance for the antiangiogenic activity of CXCL4L1 and for its potential use in clinical development but also for other biologic processes such as inflammation, thrombosis or tissue repair.
Collapse
|
38
|
Seyrek E, Dubin P. Glycosaminoglycans as polyelectrolytes. Adv Colloid Interface Sci 2010; 158:119-29. [PMID: 20444439 DOI: 10.1016/j.cis.2010.03.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2009] [Revised: 02/27/2010] [Accepted: 03/03/2010] [Indexed: 02/02/2023]
Abstract
One of the barriers to understanding structure-property relations for glycosaminoglycans has been the lack of constructive interplay between the principles and methodologies of the life sciences (molecular biology, biochemistry and cell biology) and the physical sciences, particularly in the field of polyelectrolytes. To address this, we first review the similarities and differences between the physicochemical properties of GAGs and other statistical chain polyelectrolytes of both natural and abioitic origin. Since the biofunctionality and regulation of the structures of GAGs is intimately connected with interactions with their cognate proteins, we particularly compare and contrast aspects of protein binding, i.e. effects of both GAGs and other polyelectrolytes on protein stability, protein aggregation and phase behavior. The protein binding affinities and their dependences on pH and ionic strength for the two groups are discussed not only in terms of observable differences, but also with regard to contrasting descriptions of the bound state and the role of electrostatics. We conclude that early studies of the heparin-Antithromin system, proceeding to a large extent through the methods and models of protein chemistry and drug discovery, established not only many enabling precedents but also constraining paradigms. Current studies on heparan sulfate and chondroitin sulfate seem to reflect a more ecumenical view likely to be more compatible with concepts from physical and polymer chemistry.
Collapse
Affiliation(s)
- Emek Seyrek
- CNRS, Insitut Charles Sadron, 23 Rue Loess, BP 84047, F-67037 Strasbourg 2, France
| | | |
Collapse
|
39
|
Johnstone KD, Karoli T, Liu L, Dredge K, Copeman E, Li CP, Davis K, Hammond E, Bytheway I, Kostewicz E, Chiu FCK, Shackleford DM, Charman SA, Charman WN, Harenberg J, Gonda TJ, Ferro V. Synthesis and Biological Evaluation of Polysulfated Oligosaccharide Glycosides as Inhibitors of Angiogenesis and Tumor Growth. J Med Chem 2010; 53:1686-99. [DOI: 10.1021/jm901449m] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Ken D. Johnstone
- Drug Design Group, Progen Pharmaceuticals Limited, Toowong, Queensland 4066, Australia
| | - Tomislav Karoli
- Drug Design Group, Progen Pharmaceuticals Limited, Toowong, Queensland 4066, Australia
| | - Ligong Liu
- Drug Design Group, Progen Pharmaceuticals Limited, Toowong, Queensland 4066, Australia
| | - Keith Dredge
- Drug Design Group, Progen Pharmaceuticals Limited, Toowong, Queensland 4066, Australia
| | - Elizabeth Copeman
- Drug Design Group, Progen Pharmaceuticals Limited, Toowong, Queensland 4066, Australia
| | - Cai Ping Li
- Drug Design Group, Progen Pharmaceuticals Limited, Toowong, Queensland 4066, Australia
| | - Kat Davis
- Drug Design Group, Progen Pharmaceuticals Limited, Toowong, Queensland 4066, Australia
| | - Edward Hammond
- Drug Design Group, Progen Pharmaceuticals Limited, Toowong, Queensland 4066, Australia
| | - Ian Bytheway
- Drug Design Group, Progen Pharmaceuticals Limited, Toowong, Queensland 4066, Australia
| | - Edmund Kostewicz
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Francis C. K. Chiu
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - David M. Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Susan A. Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - William N. Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Job Harenberg
- Clinical Pharmacology, Faculty of Medicine Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Thomas J. Gonda
- Molecular Oncogenesis Group, Diamantina Institute for Cancer, Immunology and Metabolic Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Vito Ferro
- Drug Design Group, Progen Pharmaceuticals Limited, Toowong, Queensland 4066, Australia
- School of Physical and Chemical Sciences, Queensland University of Technology, Brisbane, Queensland 4001, Australia
| |
Collapse
|
40
|
Rusnati M, Bugatti A, Mitola S, Leali D, Bergese P, Depero LE, Presta M. Exploiting Surface Plasmon Resonance (SPR) Technology for the Identification of Fibroblast Growth Factor-2 (FGF2) Antagonists Endowed with Antiangiogenic Activity. SENSORS (BASEL, SWITZERLAND) 2009; 9:6471-503. [PMID: 22454596 PMCID: PMC3312455 DOI: 10.3390/s90806471] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 08/17/2009] [Accepted: 08/19/2009] [Indexed: 12/31/2022]
Abstract
Angiogenesis, the process of new blood vessel formation, is implicated in various physiological/pathological conditions, including embryonic development, inflammation and tumor growth. Fibroblast growth factor-2 (FGF2) is a heparin-binding angiogenic growth factor involved in various physiopathological processes, including tumor neovascularization. Accordingly, FGF2 is considered a target for antiangiogenic therapies. Thus, numerous natural/synthetic compounds have been tested for their capacity to bind and sequester FGF2 in the extracellular environment preventing its interaction with cellular receptors. We have exploited surface plasmon resonance (SPR) technique in search for antiangiogenic FGF2 binders/antagonists. In this review we will summarize our experience in SPR-based angiogenesis research, with the aim to validate SPR as a first line screening for the identification of antiangiogenic compounds.
Collapse
Affiliation(s)
- Marco Rusnati
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Antonella Bugatti
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Stefania Mitola
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Daria Leali
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Paolo Bergese
- Chemistry for Technologies Laboratory and Department of Mechanical and Industrial Engineering, School of Engineering, University of Brescia, Brescia, 25123, Italy; E-Mails: (P.B.); (L.E.D.)
| | - Laura E. Depero
- Chemistry for Technologies Laboratory and Department of Mechanical and Industrial Engineering, School of Engineering, University of Brescia, Brescia, 25123, Italy; E-Mails: (P.B.); (L.E.D.)
| | - Marco Presta
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| |
Collapse
|
41
|
The PG500 series: novel heparan sulfate mimetics as potent angiogenesis and heparanase inhibitors for cancer therapy. Invest New Drugs 2009; 28:276-83. [PMID: 19357810 DOI: 10.1007/s10637-009-9245-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 03/16/2009] [Indexed: 10/20/2022]
Abstract
Heparan sulfate mimetics, which we have called the PG500 series, have been developed to target the inhibition of both angiogenesis and heparanase activity. This series extends the technology underpinning PI-88, a mixture of highly sulfated oligosaccharides which reached Phase III clinical development for hepatocellular carcinoma. Advances in the chemistry of the PG500 series provide numerous advantages over PI-88. These new compounds are fully sulfated, single entity oligosaccharides attached to a lipophilic moiety, which have been optimized for drug development. The rational design of these compounds has led to vast improvements in potency compared to PI-88, based on in vitro angiogenesis assays and in vivo tumor models. Based on these and other data, PG545 has been selected as the lead clinical candidate for oncology and is currently undergoing formal preclinical development as a novel treatment for advanced cancer.
Collapse
|