1
|
Gansevoort M, Wentholt S, Li Vecchi G, de Vries M, Versteeg EMM, Boekema BKHL, Choppin A, Barritault D, Chiappini F, van Kuppevelt TH, Daamen WF. Next-Generation Biomaterials for Wound Healing: Development and Evaluation of Collagen Scaffolds Functionalized with a Heparan Sulfate Mimic and Fibroblast Growth Factor 2. J Funct Biomater 2025; 16:51. [PMID: 39997585 PMCID: PMC11856099 DOI: 10.3390/jfb16020051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Fibrosis after full-thickness wound healing-especially after severe burn wounds-remains a clinically relevant problem. Biomaterials that mimic the lost dermal extracellular matrix have shown promise but cannot completely prevent scar formation. We present a novel approach where porous type I collagen scaffolds were covalently functionalized with ReGeneRating Agent (RGTA®) OTR4120. RGTA® is a glycanase-resistant heparan sulfate mimetic that promotes regeneration when applied topically to chronic wounds. OTR4120 is able to capture fibroblast growth factor 2 (FGF-2), a heparan/heparin-binding growth factor that inhibits the activity of fibrosis-driving myofibroblasts. Scaffolds with various concentrations and distributions of OTR4120 were produced. When loaded with FGF-2, collagen-OTR4120 scaffolds demonstrated sustained release of FGF-2 compared to collagen-heparin scaffolds. Their anti-fibrotic potential was investigated in vitro by seeding primary human dermal fibroblasts on the scaffolds followed by stimulation with transforming growth factor β1 (TGF-β1) to induce myofibroblast differentiation. Collagen-OTR4120(-FGF-2) scaffolds diminished the gene expression levels of several myofibroblast markers. In absence of FGF-2 the collagen-OTR4120 scaffolds displayed an inherent anti-fibrotic effect, as the expression of two fibrotic markers (TGF-β1 and type I collagen) was diminished. This work highlights the potential of collagen-OTR4120 scaffolds as biomaterials to improve skin wound healing.
Collapse
Affiliation(s)
- Merel Gansevoort
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Sabine Wentholt
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Gaia Li Vecchi
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Marjolein de Vries
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Elly M. M. Versteeg
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Bouke K. H. L. Boekema
- Burn Research Lab, Alliance of Dutch Burn Care, 1941 AJ Beverwijk, The Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | | | | | | | - Toin H. van Kuppevelt
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Willeke F. Daamen
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
2
|
Blücher RO, Lim RS, Ritchie ME, Western PS. VEGF-dependent testicular vascularisation involves MEK1/2 signalling and the essential angiogenesis factors, SOX7 and SOX17. BMC Biol 2024; 22:222. [PMID: 39354506 PMCID: PMC11445939 DOI: 10.1186/s12915-024-02003-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Abnormalities of in utero testis development are strongly associated with reproductive health conditions, including male infertility and testis cancer. In mouse testes, SOX9 and FGF9 support Sertoli cell development, while VEGF signalling is essential for the establishment of vasculature. The mitogen-activated protein kinase (MAPK) pathway is a major signalling cascade, essential for cell proliferation, differentiation and activation of Sry during primary sex-determination, but little is known about its function during fetal testis morphogenesis. We explored potential functions of MAPK signalling immediately after the establishment of testis cords in embryonic day (E)12.5 Oct4-eGFP transgenic mouse testes cultured using a MEK1/2 inhibitor. RESULTS RNA sequencing in isolated gonadal somatic cells identified 116 and 114 differentially expressed genes after 24 and 72 h of MEK1/2 inhibition, respectively. Ingenuity Pathway Analysis revealed an association of MEK1/2 signalling with biological functions such as angiogenesis, vasculogenesis and cell migration. This included a failure to upregulate the master transcriptional regulators of vascular development, Sox7 and Sox17, VEGF receptor genes, the cell adhesion factor gene Cd31 and a range of other endothelial cell markers such as Cdh5 (encoding VE-cadherin) and gap junction genes Gja4 and Gja5. In contrast, only a small number of Sertoli cell enriched genes were affected. Immunofluorescent analyses of control testes revealed that the MEK1/2 downstream target, ERK1/2 was phosphorylated in endothelial cells and Sertoli cells. Inhibition of MEK1/2 eliminated pERK1/2 in fetal testes, and CD31, VE-cadherin, SOX7 and SOX17 and endothelial cells were lost. Consistent with a role for VEGF in driving endothelial cell development in the testis, inhibition of VEGFR also abrogated pERK1/2 and SOX7 and SOX17 expressing endothelial cells. Moreover, while Sertoli cell proliferation and localisation to the testis cord basement membrane was disrupted by inhibition of MEK1/2, it was unaffected by VEGFR inhibition. Instead, inhibition of FGF signalling compromised Sertoli cell proliferation and localisation to the testis cord basement membrane. CONCLUSIONS Together, our data highlight an essential role for VEGF-dependent MEK1/2 signalling in promoting vasculature and indicate that FGF signalling through MEK1/2 regulates Sertoli cell organisation in the developing mouse testis.
Collapse
Affiliation(s)
- Rheannon O Blücher
- Centre for Reproductive Health, Hudson Institute of Medical Research and Department of Molecular and Translational Science, Monash University, Clayton, VIC, 3168, Australia
| | - Rachel S Lim
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Matthew E Ritchie
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Patrick S Western
- Centre for Reproductive Health, Hudson Institute of Medical Research and Department of Molecular and Translational Science, Monash University, Clayton, VIC, 3168, Australia.
| |
Collapse
|
3
|
Nickle A, Ko S, Merrill AE. Fibroblast growth factor 2. Differentiation 2024; 139:100733. [PMID: 37858405 PMCID: PMC11009566 DOI: 10.1016/j.diff.2023.10.001] [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: 05/22/2023] [Revised: 09/20/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023]
Abstract
Fibroblast Growth Factor 2 (FGF2), also known as basic fibroblast growth factor, is a potent stimulator of growth and differentiation in multiple tissues. Its discovery traces back over 50 years ago when it was first isolated from bovine pituitary extracts due to its ability to stimulate fibroblast proliferation. Subsequent studies investigating the genomic structure of FGF2 identified multiple protein isoforms, categorized as the low molecular weight and high molecular weight FGF2. These isoforms arise from alternative translation initiation events and exhibit unique molecular and cellular functions. In this concise review, we aim to provide an overview of what is currently known about the structure, expression, and functions of the FGF2 isoforms within the contexts of development, homeostasis, and disease.
Collapse
Affiliation(s)
- Audrey Nickle
- Center for Craniofacial Molecular Biology, Department of Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Sebastian Ko
- Center for Craniofacial Molecular Biology, Department of Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Department of Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
4
|
Pomella S, Melaiu O, Dri M, Martelli M, Gargari M, Barillari G. Effects of Angiogenic Factors on the Epithelial-to-Mesenchymal Transition and Their Impact on the Onset and Progression of Oral Squamous Cell Carcinoma: An Overview. Cells 2024; 13:1294. [PMID: 39120324 PMCID: PMC11311310 DOI: 10.3390/cells13151294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
High levels of vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2 and angiopoietin (ANG)-2 are found in tissues from oral squamous cell carcinoma (OSCC) and oral potentially malignant disorders (OPMDs). As might be expected, VEGF, FGF-2, and ANG-2 overexpression parallels the development of new blood and lymphatic vessels that nourish the growing OPMDs or OSCCs and provide the latter with metastatic routes. Notably, VEGF, FGF-2, and ANG-2 are also linked to the epithelial-to-mesenchymal transition (EMT), a trans-differentiation process that respectively promotes or exasperates the invasiveness of normal and neoplastic oral epithelial cells. Here, we have summarized published work regarding the impact that the interplay among VEGF, FGF-2, ANG-2, vessel generation, and EMT has on oral carcinogenesis. Results from the reviewed studies indicate that VEGF, FGF-2, and ANG-2 spark either protein kinase B (AKT) or mitogen-activated protein kinases (MAPK), two signaling pathways that can promote both EMT and new vessels' formation in OPMDs and OSCCs. Since EMT and vessel generation are key to the onset and progression of OSCC, as well as to its radio- and chemo-resistance, these data encourage including AKT or MAPK inhibitors and/or antiangiogenic drugs in the treatment of this malignancy.
Collapse
Affiliation(s)
- Silvia Pomella
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier, 00133 Rome, Italy; (S.P.); (O.M.); (M.M.); (M.G.)
| | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier, 00133 Rome, Italy; (S.P.); (O.M.); (M.M.); (M.G.)
| | - Maria Dri
- Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Mirko Martelli
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier, 00133 Rome, Italy; (S.P.); (O.M.); (M.M.); (M.G.)
| | - Marco Gargari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier, 00133 Rome, Italy; (S.P.); (O.M.); (M.M.); (M.G.)
| | - Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier, 00133 Rome, Italy; (S.P.); (O.M.); (M.M.); (M.G.)
| |
Collapse
|
5
|
Kulkoyluoglu Cotul E, Safdar MH, Paez SJ, Kulkarni A, Ayers MG, Lin H, Xianyu Z, Teegarden D, Hursting SD, Wendt MK. FGFR1 Signaling Facilitates Obesity-Driven Pulmonary Outgrowth in Metastatic Breast Cancer. Mol Cancer Res 2024; 22:254-267. [PMID: 38153436 PMCID: PMC10923021 DOI: 10.1158/1541-7786.mcr-23-0955] [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: 11/13/2023] [Revised: 11/30/2023] [Accepted: 12/21/2023] [Indexed: 12/29/2023]
Abstract
Survival of dormant, disseminated breast cancer cells contributes to tumor relapse and metastasis. Women with a body mass index greater than 35 have an increased risk of developing metastatic recurrence. Herein, we investigated the effect of diet-induced obesity (DIO) on primary tumor growth and metastatic progression using both metastatic and systemically dormant mouse models of breast cancer. This approach led to increased PT growth and pulmonary metastasis. We developed a novel protocol to induce obesity in Balb/c mice by combining dietary and hormonal interventions with a thermoneutral housing strategy. In contrast to standard housing conditions, ovariectomized Balb/c mice fed a high-fat diet under thermoneutral conditions became obese over a period of 10 weeks, resulting in a 250% gain in fat mass. Obese mice injected with the D2.OR model developed macroscopic pulmonary nodules compared with the dormant phenotype of these cells in mice fed a control diet. Analysis of the serum from obese Balb/c mice revealed increased levels of FGF2 as compared with lean mice. We demonstrate that serum from obese animals, exogenous FGF stimulation, or constitutive stimulation through autocrine and paracrine FGF2 is sufficient to break dormancy and drive pulmonary outgrowth. Blockade of FGFR signaling or specific depletion of FGFR1 prevented obesity-associated outgrowth of the D2.OR model. IMPLICATIONS Overall, this study developed a novel DIO model that allowed for demonstration of FGF2:FGFR1 signaling as a key molecular mechanism connecting obesity to breakage of systemic tumor dormancy and metastatic progression.
Collapse
Affiliation(s)
- Eylem Kulkoyluoglu Cotul
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Muhammad Hassan Safdar
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Sebastian Juan Paez
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Aneesha Kulkarni
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Mitchell G. Ayers
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Hang Lin
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Zilin Xianyu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Dorothy Teegarden
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, IN, United States
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
| | - Stephen D. Hursting
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael K. Wendt
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
6
|
Kolkmann AM, Van Essen A, Post MJ, Moutsatsou P. Development of a Chemically Defined Medium for in vitro Expansion of Primary Bovine Satellite Cells. Front Bioeng Biotechnol 2022; 10:895289. [PMID: 35992337 PMCID: PMC9385969 DOI: 10.3389/fbioe.2022.895289] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 04/18/2022] [Indexed: 11/19/2022] Open
Abstract
The use of fetal bovine serum (FBS) in animal cell culture media is widely spread since it provides a broad spectrum of molecules that are known to support cell attachment and growth. However, the harvest and collection procedures of FBS raise ethical concerns and serum is an ill-defined and expensive component. This is especially problematic when it comes to regulatory approval for food applications like cultured meat. The aim of this study is to develop a chemically defined, cost efficient serum-free and animal-free medium that supports the attachment and expansion of bovine myoblasts while maintaining their differentiation capacity. Bovine satellite cells were harvested and isolated from a fresh sample of skeletal muscle tissue and cultured in planar systems. The efficacy of the tested formulations was assessed with metabolic assays and cell counting techniques. Optical microscopy was used to observe cellular morphology and statistical analysis was applied. Based on a comprehensive literature analysis, a defined serum-free medium (SFM) composition was developed consisting of DMEM/F12 as basal medium, supplemented with L-ascorbic acid 2-phosphate, fibronectin, hydrocortisone, GlutaMAX™, albumin, ITS-X, hIL-6, α-linolenic acid, and growth factors such as FGF-2, VEGF, IGF-1, HGF, and PDGF-BB. To our knowledge, this is the first defined serum-free and animal free medium formulation specific for bovine myoblasts to date. We conclude that the SFM formulation supported exponential cell growth up to 97% of the serum-containing golden standard growth medium. All reagents used in this study are chemically defined.
Collapse
Affiliation(s)
- Anna M. Kolkmann
- Mosa Meat BV, Maastricht, Netherlands
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| | | | - Mark J. Post
- Mosa Meat BV, Maastricht, Netherlands
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| | - Panagiota Moutsatsou
- Mosa Meat BV, Maastricht, Netherlands
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
7
|
Yang Z, Chen S, Ying H, Yao W. Targeting syndecan-1: new opportunities in cancer therapy. Am J Physiol Cell Physiol 2022; 323:C29-C45. [PMID: 35584326 PMCID: PMC9236862 DOI: 10.1152/ajpcell.00024.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 12/02/2022]
Abstract
Syndecan-1 (SDC1, CD138) is one of the heparan sulfate proteoglycans and is essential for maintaining normal cell morphology, interacting with the extracellular and intracellular protein repertoire, as well as mediating signaling transduction upon environmental stimuli. The critical role of SDC1 in promoting tumorigenesis and metastasis has been increasingly recognized in various cancer types, implying a promising potential of utilizing SDC1 as a novel target for cancer therapy. This review summarizes the current knowledge on SDC1 structure and functions, including its role in tumor biology. We also discuss the highlights and limitations of current SDC1-targeted therapies as well as the obstacles in developing new therapeutic methods, offering our perspective on the future directions to target SDC1 for cancer treatment.
Collapse
Affiliation(s)
- Zecheng Yang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shuaitong Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wantong Yao
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
8
|
Yang H, Liu Y, Zhao MM, Guo Q, Zheng XK, Liu D, Zeng KW, Tu PF. Therapeutic potential of targeting membrane-spanning proteoglycan SDC4 in hepatocellular carcinoma. Cell Death Dis 2021; 12:492. [PMID: 33990545 PMCID: PMC8121893 DOI: 10.1038/s41419-021-03780-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022]
Abstract
Syndecan-4 (SDC4) functions as a major endogenous membrane-associated receptor and widely regulates cytoskeleton, cell adhesion, and cell migration in human tumorigenesis and development, which represents a charming anti-cancer therapeutic target. Here, SDC4 was identified as a direct cellular target of small-molecule bufalin with anti-hepatocellular carcinoma (HCC) activity. Mechanism studies revealed that bufalin directly bond to SDC4 and selectively increased SDC4 interaction with substrate protein DEAD-box helicase 23 (DDX23) to induce HCC genomic instability. Meanwhile, pharmacological promotion of SDC4/DDX23 complex formation also inactivated matrix metalloproteinases (MMPs) and augmented p38/JNK MAPKs phosphorylation, which are highly associated with HCC proliferation and migration. Notably, specific knockdown of SDC4 or DDX23 markedly abolished bufalin-dependent inhibition of HCC proliferation and migration, indicating SDC4/DDX23 signaling axis is highly involved in the HCC process. Our results indicate that membrane-spanning proteoglycan SDC4 is a promising druggable target for HCC, and pharmacological regulation of SDC4/DDX23 signaling axis with small-molecule holds great potential to benefit HCC patients.
Collapse
Affiliation(s)
- Heng Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yang Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Mei-Mei Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Qiang Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xi-Kang Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Dan Liu
- Proteomics Laboratory, Medical and Healthy Analytical Center, Peking University Health Science Center, Beijing, 100191, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Peng-Fei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
9
|
Heparan Sulfate Deficiency in Cartilage: Enhanced BMP-Sensitivity, Proteoglycan Production and an Anti-Apoptotic Expression Signature after Loading. Int J Mol Sci 2021; 22:ijms22073726. [PMID: 33918436 PMCID: PMC8038223 DOI: 10.3390/ijms22073726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis (OA) represents one major cause of disability worldwide still evading efficient pharmacological or cellular therapies. Severe degeneration of extracellular cartilage matrix precedes the loss of mobility and disabling pain perception in affected joints. Recent studies showed that a reduced heparan sulfate (HS) content protects cartilage from degradation in OA-animal models of joint destabilization but the underlying mechanisms remained unclear. We aimed to clarify whether low HS-content alters the mechano-response of chondrocytes and to uncover pathways relevant for HS-related chondro-protection in response to loading. Tissue-engineered cartilage with HS-deficiency was generated from rib chondrocytes of mice carrying a hypomorphic allele of Exostosin 1 (Ext1), one of the main HS-synthesizing enzymes, and wildtype (WT) littermate controls. Engineered cartilage matured for 2 weeks was exposed to cyclic unconfined compression in a bioreactor. The molecular loading response was determined by transcriptome profiling, bioinformatic data processing, and qPCR. HS-deficient chondrocytes expressed 3-6% of WT Ext1-mRNA levels. Both groups similarly raised Sox9, Col2a1 and Acan levels during maturation. However, HS-deficient chondrocytes synthesized and deposited 50% more GAG/DNA. TGFβ and FGF2-sensitivity of Ext1gt/gt chondrocytes was similar to WT cells but their response to BMP-stimulation was enhanced. Loading induced similar activation of mechano-sensitive ERK and P38-signaling in WT and HS-reduced chondrocytes. Transcriptome analysis reflected regulation of cell migration as major load-induced biological process with similar stimulation of common (Fosl1, Itgα5, Timp1, and Ngf) as well as novel mechano-regulated genes (Inhba and Dhrs9). Remarkably, only Ext1-hypomorphic cartilage responded to loading by an expression signature of negative regulation of apoptosis with pro-apoptotic Bnip3 being selectively down-regulated. HS-deficiency enhanced BMP-sensitivity, GAG-production and fostered an anti-apoptotic expression signature after loading, all of which may protect cartilage from load-induced erosion.
Collapse
|
10
|
Burgos-Bravo F, Martínez-Meza S, Quest AFG, Wilson CAM, Leyton L. Application of Force to a Syndecan-4 Containing Complex With Thy-1-α Vβ 3 Integrin Accelerates Neurite Retraction. Front Mol Biosci 2020; 7:582257. [PMID: 33134319 PMCID: PMC7550751 DOI: 10.3389/fmolb.2020.582257] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 08/25/2020] [Indexed: 12/23/2022] Open
Abstract
Inflammation contributes to the genesis and progression of chronic diseases, such as cancer and neurodegeneration. Upregulation of integrins in astrocytes during inflammation induces neurite retraction by binding to the neuronal protein Thy-1, also known as CD90. Additionally, Thy-1 alters astrocyte contractility and movement by binding to the mechano-sensors αVβ3 integrin and Syndecan-4. However, the contribution of Syndecan-4 to neurite shortening following Thy-1-αVβ3 integrin interaction remains unknown. To further characterize the contribution of Syndecan-4 in Thy-1-dependent neurite outgrowth inhibition and neurite retraction, cell-based assays under pro-inflammatory conditions were performed. In addition, using Optical Tweezers, we studied single-molecule binding properties between these proteins, and their mechanical responses. Syndecan-4 increased the lifetime of Thy-1-αVβ3 integrin binding by interacting directly with Thy-1 and forming a ternary complex (Thy-1-αVβ3 integrin + Syndecan-4). Under in vitro-generated pro-inflammatory conditions, Syndecan-4 accelerated the effect of integrin-engaged Thy-1 by forming this ternary complex, leading to faster neurite retraction and the inhibition of neurite outgrowth. Thus, Syndecan-4 controls neurite cytoskeleton contractility by modulating αVβ3 integrin mechano-receptor function. These results suggest that mechano-transduction, cell-matrix and cell-cell interactions are likely critical events in inflammation-related disease development.
Collapse
Affiliation(s)
- Francesca Burgos-Bravo
- Laboratory of Cellular Communication, Center for Studies on Exercise, Metabolism and Cancer, Institute of Biomedical Sciences, Santiago, Chile.,Advanced Center for Chronic Diseases, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Single Molecule Biochemistry and Mechanobiology Laboratory, Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Samuel Martínez-Meza
- Laboratory of Cellular Communication, Center for Studies on Exercise, Metabolism and Cancer, Institute of Biomedical Sciences, Santiago, Chile.,Advanced Center for Chronic Diseases, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Andrew F G Quest
- Laboratory of Cellular Communication, Center for Studies on Exercise, Metabolism and Cancer, Institute of Biomedical Sciences, Santiago, Chile.,Advanced Center for Chronic Diseases, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Christian A M Wilson
- Single Molecule Biochemistry and Mechanobiology Laboratory, Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Lisette Leyton
- Laboratory of Cellular Communication, Center for Studies on Exercise, Metabolism and Cancer, Institute of Biomedical Sciences, Santiago, Chile.,Advanced Center for Chronic Diseases, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
11
|
Xiong A, Spyrou A, Forsberg-Nilsson K. Involvement of Heparan Sulfate and Heparanase in Neural Development and Pathogenesis of Brain Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:365-403. [PMID: 32274718 DOI: 10.1007/978-3-030-34521-1_14] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Brain tumors are aggressive and devastating diseases. The most common type of brain tumor, glioblastoma (GBM), is incurable and has one of the worst five-year survival rates of all human cancers. GBMs are invasive and infiltrate healthy brain tissue, which is one main reason they remain fatal despite resection, since cells that have already migrated away lead to rapid regrowth of the tumor. Curative therapy for medulloblastoma (MB), the most common pediatric brain tumor, has improved, but the outcome is still poor for many patients, and treatment causes long-term complications. Recent advances in the classification of pediatric brain tumors reveal distinct subgroups, allowing more targeted therapy for the most aggressive forms, and sparing children with less malignant tumors the side-effects of massive treatment. Heparan sulfate proteoglycans (HSPGs), main components of the neurogenic niche, interact specifically with a large number of physiologically important molecules and vital roles for HS biosynthesis and degradation in neural stem cell differentiation have been presented. HSPGs are composed of a core protein with attached highly charged, sulfated disaccharide chains. The major enzyme that degrades HS is heparanase (HPSE), an important regulator of extracellular matrix (ECM) remodeling which has been suggested to promote the growth and invasion of other types of tumors. This is of clinical interest because GBM are highly invasive and children with metastatic MB at the time of diagnosis exhibit a worse outcome. Here we review the involvement of HS and HPSE in development of the nervous system and some of its most malignant brain tumors, glioblastoma and medulloblastoma.
Collapse
Affiliation(s)
- Anqi Xiong
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Insitutet, Stockholm, Sweden
| | - Argyris Spyrou
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
12
|
Vitorino FNDL, Montoni F, Moreno JN, de Souza BF, Lopes MDC, Cordeiro B, Fonseca CS, Gilmore JM, Sardiu MI, Reis MS, Florens LA, Washburn MP, Armelin HA, da Cunha JPC. FGF2 Antiproliferative Stimulation Induces Proteomic Dynamic Changes and High Expression of FOSB and JUNB in K-Ras-Driven Mouse Tumor Cells. Proteomics 2018; 18:e1800203. [PMID: 30035358 DOI: 10.1002/pmic.201800203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/28/2018] [Indexed: 11/07/2022]
Abstract
Fibroblast growth factor 2 (FGF2) is a well-known cell proliferation promoter; however, it can also induce cell cycle arrest. To gain insight into the molecular mechanisms of this antiproliferative effect, for the first time, the early systemic proteomic differences induced by this growth factor in a K-Ras-driven mouse tumor cell line using a quantitative proteomics approach are investigated. More than 2900 proteins are quantified, indicating that terms associated with metabolism, RNA processing, replication, and transcription are enriched among proteins differentially expressed upon FGF2 stimulation. Proteomic trend dynamics indicate that, for proteins mainly associated with DNA replication and carbohydrate metabolism, an FGF2 stimulus delays their abundance changes, whereas FGF2 stimulation accelerates other metabolic programs. Transcription regulatory network analysis indicates master regulators of FGF2 stimulation, including two critical transcription factors, FOSB and JUNB. Their expression dynamics, both in the Y1 cell line (a murine model of adenocarcinoma cells) and in two other human cell lines (SK-N-MC and UM-UC-3) also susceptible to FGF2 antiproliferative effects, are investigated. Both protein expression levels depend on fibroblast growth factor receptor (FGFR) and src signaling. JUNB and FOSB knockdown do not rescue cells from the growth arrest induced by FGF2; however, FOSB knockdown rescue cells from DNA replication delay, indicating that FOSB expression underlies one of the FGF2 antiproliferative effects, namely, S-phase progression delay.
Collapse
Affiliation(s)
- Francisca Nathalia de Luna Vitorino
- Laboratório Especial de Ciclo Celular - Center of Toxins, Immune-Response and Cell Signaling - CeTICS, Instituto Butantan, São Paulo, SP, 05503-900, Brazil
| | - Fabio Montoni
- Laboratório Especial de Ciclo Celular - Center of Toxins, Immune-Response and Cell Signaling - CeTICS, Instituto Butantan, São Paulo, SP, 05503-900, Brazil
| | - Jaqueline Neves Moreno
- Laboratório Especial de Ciclo Celular - Center of Toxins, Immune-Response and Cell Signaling - CeTICS, Instituto Butantan, São Paulo, SP, 05503-900, Brazil
| | - Bruno Ferreira de Souza
- Laboratório Especial de Ciclo Celular - Center of Toxins, Immune-Response and Cell Signaling - CeTICS, Instituto Butantan, São Paulo, SP, 05503-900, Brazil
| | - Mariana de Camargo Lopes
- Laboratório Especial de Ciclo Celular - Center of Toxins, Immune-Response and Cell Signaling - CeTICS, Instituto Butantan, São Paulo, SP, 05503-900, Brazil
| | - Barbara Cordeiro
- Laboratório Especial de Ciclo Celular - Center of Toxins, Immune-Response and Cell Signaling - CeTICS, Instituto Butantan, São Paulo, SP, 05503-900, Brazil
| | - Cecilia Sella Fonseca
- Laboratório Especial de Ciclo Celular - Center of Toxins, Immune-Response and Cell Signaling - CeTICS, Instituto Butantan, São Paulo, SP, 05503-900, Brazil
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Joshua M Gilmore
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Mihaela I Sardiu
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Marcelo Silva Reis
- Laboratório Especial de Ciclo Celular - Center of Toxins, Immune-Response and Cell Signaling - CeTICS, Instituto Butantan, São Paulo, SP, 05503-900, Brazil
| | | | - Michael P Washburn
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66045, USA
| | - Hugo Aguirre Armelin
- Laboratório Especial de Ciclo Celular - Center of Toxins, Immune-Response and Cell Signaling - CeTICS, Instituto Butantan, São Paulo, SP, 05503-900, Brazil
| | - Julia Pinheiro Chagas da Cunha
- Laboratório Especial de Ciclo Celular - Center of Toxins, Immune-Response and Cell Signaling - CeTICS, Instituto Butantan, São Paulo, SP, 05503-900, Brazil
| |
Collapse
|
13
|
Nguyen EH, Murphy WL. Customizable biomaterials as tools for advanced anti-angiogenic drug discovery. Biomaterials 2018; 181:53-66. [PMID: 30077137 DOI: 10.1016/j.biomaterials.2018.07.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/17/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
The inhibition of angiogenesis is a critical element of cancer therapy, as cancer vasculature contributes to tumor expansion. While numerous drugs have proven to be effective at disrupting cancer vasculature, patient survival has not significantly improved as a result of anti-angiogenic drug treatment. Emerging evidence suggests that this is due to a combination of unintended side effects resulting from the application of anti-angiogenic compounds, including angiogenic rebound after treatment and the activation of metastasis in the tumor. There is currently a need to better understand the far-reaching effects of anti-angiogenic drug treatments in the context of cancer. Numerous innovations and discoveries in biomaterials design and tissue engineering techniques are providing investigators with tools to develop physiologically relevant vascular models and gain insights into the holistic impact of drug treatments on tumors. This review examines recent advances in the design of pro-angiogenic biomaterials, specifically in controlling integrin-mediated cell adhesion, growth factor signaling, mechanical properties and oxygen tension, as well as the implementation of pro-angiogenic materials into sophisticated co-culture models of cancer vasculature.
Collapse
Affiliation(s)
- Eric H Nguyen
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Human Models for Analysis of Pathways (Human MAPs) Center, University of Wisconsin, Madison, WI, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Human Models for Analysis of Pathways (Human MAPs) Center, University of Wisconsin, Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
14
|
Guo M, Shi JH, Wang PL, Shi DZ. Angiogenic Growth Factors for Coronary Artery Disease: Current Status and Prospects. J Cardiovasc Pharmacol Ther 2017; 23:130-141. [PMID: 29025278 DOI: 10.1177/1074248417735399] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Ming Guo
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun-He Shi
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL, USA
| | - Pei-Li Wang
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Da-Zhuo Shi
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Matsumoto K, Ayukawa T, Ishio A, Sasamura T, Yamakawa T, Matsuno K. Dual Roles of O-Glucose Glycans Redundant with Monosaccharide O-Fucose on Notch in Notch Trafficking. J Biol Chem 2016; 291:13743-52. [PMID: 27129198 DOI: 10.1074/jbc.m115.710483] [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: 12/17/2015] [Indexed: 12/25/2022] Open
Abstract
Notch is a transmembrane receptor that mediates cell-cell interactions and controls various cell-fate specifications in metazoans. The extracellular domain of Notch contains multiple epidermal growth factor (EGF)-like repeats. At least five different glycans are found in distinct sites within these EGF-like repeats. The function of these individual glycans in Notch signaling has been investigated, primarily by disrupting their individual glycosyltransferases. However, we are just beginning to understand the potential functional interactions between these glycans. Monosaccharide O-fucose and O-glucose trisaccharide (O-glucose-xylose-xylose) are added to many of the Notch EGF-like repeats. In Drosophila, Shams adds a xylose specifically to the monosaccharide O-glucose. We found that loss of the terminal dixylose of O-glucose-linked saccharides had little effect on Notch signaling. However, our analyses of double mutants of shams and other genes required for glycan modifications revealed that both the monosaccharide O-glucose and the terminal dixylose of O-glucose-linked saccharides function redundantly with the monosaccharide O-fucose in Notch activation and trafficking. The terminal dixylose of O-glucose-linked saccharides and the monosaccharide O-glucose were required in distinct Notch trafficking processes: Notch transport from the apical plasma membrane to adherens junctions, and Notch export from the endoplasmic reticulum, respectively. Therefore, the monosaccharide O-glucose and terminal dixylose of O-glucose-linked saccharides have distinct activities in Notch trafficking, although a loss of these activities is compensated for by the presence of monosaccharide O-fucose. Given that various glycans attached to a protein motif may have redundant functions, our results suggest that these potential redundancies may lead to a serious underestimation of glycan functions.
Collapse
Affiliation(s)
- Kenjiroo Matsumoto
- From the Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043 and
| | - Tomonori Ayukawa
- the Department of Biological Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-1500, Japan
| | - Akira Ishio
- the Department of Biological Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-1500, Japan
| | - Takeshi Sasamura
- From the Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043 and
| | - Tomoko Yamakawa
- From the Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043 and
| | - Kenji Matsuno
- From the Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043 and
| |
Collapse
|
16
|
Akl MR, Nagpal P, Ayoub NM, Prabhu SA, Gliksman M, Tai B, Hatipoglu A, Goy A, Suh KS. Molecular and clinical profiles of syndecan-1 in solid and hematological cancer for prognosis and precision medicine. Oncotarget 2015; 6:28693-715. [PMID: 26293675 PMCID: PMC4745686 DOI: 10.18632/oncotarget.4981] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/11/2015] [Indexed: 12/18/2022] Open
Abstract
Syndecan-1 (SDC1, CD138) is a key cell surface adhesion molecule essential for maintaining cell morphology and interaction with the surrounding microenvironment. Deregulation of SDC1 contributes to cancer progression by promoting cell proliferation, metastasis, invasion and angiogenesis, and is associated with relapse through chemoresistance. SDC1 expression level is also associated with responses to chemotherapy and with prognosis in multiple solid and hematological cancers, including multiple myeloma and Hodgkin lymphoma. At the tissue level, the expression levels of SDC1 and the released extracellular domain of SDC1 correlate with tumor malignancy, phenotype, and metastatic potential for both solid and hematological tumors in a tissue-specific manner. The SDC1 expression profile varies among cancer types, but the differential expression signatures between normal and cancer cells in epithelial and stromal compartments are directly associated with aggressiveness of tumors and patient's clinical outcome and survival. Therefore, relevant biomarkers of SDC signaling may be useful for selecting patients that would most likely respond to a particular therapy at the time of diagnosis or perhaps for predicting relapse. In addition, the reciprocal expression signature of SDC between tumor epithelial and stromal compartments may have synergistic value for patient selection and the prediction of clinical outcome.
Collapse
Affiliation(s)
- Mohamed R. Akl
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Poonam Nagpal
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Sathyen A. Prabhu
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Matthew Gliksman
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Betty Tai
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Ahmet Hatipoglu
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Andre Goy
- Lymphoma Division, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - K. Stephen Suh
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| |
Collapse
|
17
|
Teran M, Nugent MA. Synergistic Binding of Vascular Endothelial Growth Factor-A and Its Receptors to Heparin Selectively Modulates Complex Affinity. J Biol Chem 2015; 290:16451-62. [PMID: 25979342 DOI: 10.1074/jbc.m114.627372] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Indexed: 01/13/2023] Open
Abstract
Angiogenesis is a highly regulated process orchestrated by the VEGF system. Heparin/heparan sulfate proteoglycans and neuropilin-1 (NRP-1) have been identified as co-receptors, yet the mechanisms of action have not been fully defined. In the present study, we characterized molecular interactions between receptors and co-receptors, using surface plasmon resonance and in vitro binding assays. Additionally, we demonstrate that these binding events are relevant to VEGF activity within endothelial cells. We defined interactions and structural requirements for heparin/HS interactions with VEGF receptor (VEGFR)-1, NRP-1, and VEGF165 in complex with VEGFR-2 and NRP-1. We demonstrate that these structural requirements are distinct for each interaction. We further show that VEGF165, VEGFR-2, and monomeric NRP-1 bind weakly to heparin alone yet show synergistic binding to heparin when presented together in various combinations. This synergistic binding appears to translate to alterations in VEGF signaling in endothelial cells. We found that soluble NRP-1 increases VEGF binding and activation of VEGFR-2 and ERK1/2 in endothelial cells and that these effects require sulfated HS. These data suggest that the presence of HS/heparin and NRP-1 may dictate the specific receptor type activated by VEGF and ultimately determine the biological output of the system. The ability of co-receptors to fine-tune VEGF responsiveness suggests the possibility that VEGF-mediated angiogenesis can be selectively stimulated or inhibited by targeting HS/heparin and NRP-1.
Collapse
Affiliation(s)
- Madelane Teran
- From the Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118 and
| | - Matthew A Nugent
- the Department of Biological Sciences, University of Massachusetts Lowell, Lowell, Massachusetts 01854
| |
Collapse
|
18
|
Abstract
The vascular network carries blood throughout the body, delivering oxygen to tissues and providing a pathway for communication between distant organs. The network is hierarchical and structured, but also dynamic, especially at the smaller scales. Remodeling of the microvasculature occurs in response to local changes in oxygen, gene expression, cell-cell communication, and chemical and mechanical stimuli from the microenvironment. These local changes occur as a result of physiological processes such as growth and exercise, as well as acute and chronic diseases including stroke, cancer, and diabetes, and pharmacological intervention. While the vasculature is an important therapeutic target in many diseases, drugs designed to inhibit vascular growth have achieved only limited success, and no drug has yet been approved to promote therapeutic vascular remodeling. This highlights the challenges involved in identifying appropriate therapeutic targets in a system as complex as the vasculature. Systems biology approaches provide a means to bridge current understanding of the vascular system, from detailed signaling dynamics measured in vitro and pre-clinical animal models of vascular disease, to a more complete picture of vascular regulation in vivo. This will translate to an improved ability to identify multi-component biomarkers for diagnosis, prognosis, and monitoring of therapy that are easy to measure in vivo, as well as better drug targets for specific disease states. In this review, we summarize systems biology approaches that have advanced our understanding of vascular function and dysfunction in vivo, with a focus on computational modeling.
Collapse
Affiliation(s)
- Lindsay E Clegg
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| | | |
Collapse
|
19
|
Wang P, Zhang L, Yao J, Shi Y, Li P, Ding K. An arabinogalactan from flowers of Panax notoginseng inhibits angiogenesis by BMP2/Smad/Id1 signaling. Carbohydr Polym 2015; 121:328-35. [PMID: 25659706 DOI: 10.1016/j.carbpol.2014.11.073] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/26/2014] [Accepted: 11/29/2014] [Indexed: 11/25/2022]
Abstract
Angiogenesis plays an essential role in tumor development. Blocking angiogenesis in tumor has become a promising tactic in limiting cancer progression. Here, an arabinogalactan polysaccharide, RN1 was isolated from flowers of Panax notoginseng. Its structure was determined to possess a backbone of 1,6-linked Galp branched at C3 by side 1,3-linked Galp, with branches attached at position O-3 of it. The branches mainly contained 1,5-linked, 1,3,5-linked, terminal Arabinose and terminal Galactose. RN1 could inhibit microvessel formation in the BxPC-3 pancreatic cancer cell xenograft tumor in nude mice. The antiangiogenesis assay showed that RN1 could reduce the migratory activity of endothelial cells and their ability of tube formation on matrigel, but no effect on endothelial cells growth. Further studies revealed that RN1 could inhibit BMP2/Smad1/5/8/Id1 signaling. All those data indicated the RN1 had an antiangiogenic effect via BMP2 signaling and could be a potential novel inhibitor of angiogenesis.
Collapse
Affiliation(s)
- Peipei Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Glycochemistry & Glycobiology Lab, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lei Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Glycochemistry & Glycobiology Lab, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jian Yao
- Glycochemistry & Glycobiology Lab, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yikang Shi
- National Glycoengineering Research Center, Shandong University, Jinan, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Kan Ding
- Glycochemistry & Glycobiology Lab, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
20
|
Oh JS, Youm YS, Cho SD, Choi SW, Cho YJ. The expression of vascular endothelial growth factor and Syndecan-4 in cartilage from osteoarthritic knees. Bone Joint J 2014; 96-B:1319-24. [PMID: 25274915 DOI: 10.1302/0301-620x.96b10.33812] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Previous studies support the important role of vascular endothelial growth factor (VEGF) and syndecan-4 in the pathogenesis of osteoarthritis (OA). Both VEGF and syndecan-4 are expressed by chondrocytes and both are involved in the regulation of matrix metalloproteinase-3, resulting in the activation of aggrecanase II (ADAMTS-5), which is essential in the pathogenesis of OA. However, the relationship between VEGF and syndecan-4 has not been established. As a pilot study, we assayed the expression of VEGF and syndecan-4 in cartilage samples and cultured chondrocytes from osteoarthritic knee joints and analysed the relationship between these two factors. Specimens were collected from 21 female patients (29 knees) who underwent total knee replacement due to severe medial OA of the knee (Kellgren-Lawrence grade 4). Articular cartilage samples, obtained from bone and cartilage excised during surgery, were analysed and used for chondrocyte culture. We found that the levels of expression of VEGF and syndecan-4 mRNA did not differ significantly between medial femoral cartilage with severe degenerative changes and lateral femoral cartilage that appeared grossly normal (p = 0.443 and 0.622, respectively). Likewise, the levels of expression of VEGF and syndecan-4 mRNA were similar in cultured chondrocytes from medial and lateral femoral cartilage. The levels of expression of VEGF and syndecan-4 mRNAs were significantly and positively correlated in cartilage explant (r = 0.601, p = 0.003) but not in cultured chondrocytes. These results suggest that there is a close relationship between VEGF and syndecan-4 in the cartilage of patients with OA. Further studies are needed to determine the exact pathway by which these two factors interact in the pathogenesis of OA.
Collapse
Affiliation(s)
- J S Oh
- University of Ulsan College of Medicine, Department of Orthopedic Surgery, Ulsan University Hospital, 290-3 Jeonha-dong, Dong-gu, Ulsan 682-714, Korea
| | - Y S Youm
- University of Ulsan College of Medicine, Department of Orthopedic Surgery, Ulsan University Hospital, 290-3 Jeonha-dong, Dong-gu, Ulsan 682-714, Korea
| | - S D Cho
- University of Ulsan College of Medicine, Department of Orthopedic Surgery, Ulsan University Hospital, 290-3 Jeonha-dong, Dong-gu, Ulsan 682-714, Korea
| | - S W Choi
- University of Ulsan College of Medicine, Department of Orthopedic Surgery, Ulsan University Hospital, 290-3 Jeonha-dong, Dong-gu, Ulsan 682-714, Korea
| | - Y J Cho
- University of Ulsan College of Medicine, Department of Orthopedic Surgery, Ulsan University Hospital, 290-3 Jeonha-dong, Dong-gu, Ulsan 682-714, Korea
| |
Collapse
|
21
|
Xiong A, Kundu S, Forsberg-Nilsson K. Heparan sulfate in the regulation of neural differentiation and glioma development. FEBS J 2014; 281:4993-5008. [DOI: 10.1111/febs.13097] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 09/17/2014] [Accepted: 10/02/2014] [Indexed: 12/20/2022]
Affiliation(s)
- Anqi Xiong
- Department of Immunology, Genetics and Pathology, and Science for Life Laboratory; Rudbeck Laboratory; Uppsala University; Uppsala Sweden
| | - Soumi Kundu
- Department of Immunology, Genetics and Pathology, and Science for Life Laboratory; Rudbeck Laboratory; Uppsala University; Uppsala Sweden
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology, and Science for Life Laboratory; Rudbeck Laboratory; Uppsala University; Uppsala Sweden
| |
Collapse
|
22
|
Fujisawa C, Castellot JJ. Matrix production and remodeling as therapeutic targets for uterine leiomyoma. J Cell Commun Signal 2014; 8:179-94. [PMID: 25012731 DOI: 10.1007/s12079-014-0234-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 05/22/2014] [Indexed: 01/06/2023] Open
Abstract
Uterine leiomyoma, commonly known as fibroids, is a benign neoplasm of smooth muscle in women. The incidence of clinically symptomatic fibroids in reproductive-age women is approximately 20 %, with nearly 80 % of black women suffering from this condition. Symptoms include severe pain and hemorrhage; fibroids are also a major cause of infertility or sub-fertility in women. Uterine leiomyoma consist of hyperplastic smooth muscle cells and an excess deposition of extracellular matrix, specifically collagen, fibronectin, and sulfated proteoglycans. Extracellular matrix components interact and signal through integrin-β1 on the surface of uterine leiomyoma smooth muscle cells, provide growth factor storage, and act as co-receptors for growth factor-receptor binding. ECM and growth factor signaling through integrin-β1 and growth factor receptors significantly increases cell proliferation and ECM deposition in uterine leiomyoma. Growth factors TGF-β, IGF, PDGF, FGF and EGF are all shown to promote uterine leiomyoma progression and signal through multiple pathways to increase the expression of genes encoding matrix or matrix-modifying proteins. Decreasing integrin expression, reducing growth factor action and inhibiting ECM action on uterine leiomyoma smooth muscle cells are important opportunities to treat uterine leiomyoma without use of the current surgical procedures. Both natural compounds and chemicals are shown to decrease fibrosis and uterine leiomyoma progression, but further analysis is needed to make inroads in treating this common women's health issue.
Collapse
Affiliation(s)
- Caitlin Fujisawa
- Public Heath and Professional Degrees Program, Tufts University School of Medicine, Boston, MA, 02111, USA
| | | |
Collapse
|
23
|
Kanodia J, Chai D, Vollmer J, Kim J, Raue A, Finn G, Schoeberl B. Deciphering the mechanism behind Fibroblast Growth Factor (FGF) induced biphasic signal-response profiles. Cell Commun Signal 2014; 12:34. [PMID: 24885272 PMCID: PMC4036111 DOI: 10.1186/1478-811x-12-34] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 04/28/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The Fibroblast Growth Factor (FGF) pathway is driving various aspects of cellular responses in both normal and malignant cells. One interesting characteristic of this pathway is the biphasic nature of the cellular response to some FGF ligands like FGF2. Specifically, it has been shown that phenotypic behaviors controlled by FGF signaling, like migration and growth, reach maximal levels in response to intermediate concentrations, while high levels of FGF2 elicit weak responses. The mechanisms leading to the observed biphasic response remains unexplained. RESULTS A combination of experiments and computational modeling was used to understand the mechanism behind the observed biphasic signaling responses. FGF signaling involves a tertiary surface interaction that we captured with a computational model based on Ordinary Differential Equations (ODEs). It accounts for FGF2 binding to FGF receptors (FGFRs) and heparan sulfate glycosaminoglycans (HSGAGs), followed by receptor-phosphorylation, activation of the FRS2 adapter protein and the Ras-Raf signaling cascade. Quantitative protein assays were used to measure the dynamics of phosphorylated ERK (pERK) in response to a wide range of FGF2 ligand concentrations on a fine-grained time scale for the squamous cell lung cancer cell line H1703. We developed a novel approach combining Particle Swarm Optimization (PSO) and feature-based constraints in the objective function to calibrate the computational model to the experimental data. The model is validated using a series of extracellular and intracellular perturbation experiments. We demonstrate that in silico model predictions are in accordance with the observed in vitro results. CONCLUSIONS Using a combined approach of computational modeling and experiments we found that competition between binding of the ligand FGF2 to HSGAG and FGF receptor leads to the biphasic response. At low to intermediate concentrations of FGF2 there are sufficient free FGF receptors available for the FGF2-HSGAG complex to enable the formation of the trimeric signaling unit. At high ligand concentrations the ligand binding sites of the receptor become saturated and the trimeric signaling unit cannot be formed. This insight into the pathway is an important consideration for the pharmacological inhibition of this pathway.
Collapse
Affiliation(s)
- Jitendra Kanodia
- Merrimack Pharmaceuticals, Suite B7201, 1 Kendall Square, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Datta P, Yang B, Linhardt RJ, Sharfstein ST. Modulation of heparan sulfate biosynthesis by sodium butyrate in recombinant CHO cells. Cytotechnology 2014; 67:223-35. [PMID: 24468831 DOI: 10.1007/s10616-013-9677-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 12/14/2013] [Indexed: 12/19/2022] Open
Abstract
Sodium butyrate, a histone deacetylase inhibitor, has been used to improve transgene expression in Chinese hamster ovary (CHO) cells. The current study explores the impact of butyrate treatment on heparan sulfate (HS) biosynthesis and structural composition in a recombinant CHO-S cell line expressing enzymes in the heparin (HP)/(HS) biosynthetic pathway (Dual-10 stably expressing NDST2 and HS3st1). Flow cytometric analysis showed that antithrombin binding was increased in Dual-10 cells and basic fibroblast growth factor binding was decreased in response to sodium butyrate treatment. The results were in agreement with the AMAC-LCMS (2-aminoacridine-tagged HS/HP analysis by liquid chromatography mass spectrometry) data that showed that there was an increase in heparan sulfate tri-sulfated disaccharides and a decrease in N-sulfated disaccharides in the butyrate-treated cells. However, we could not detect any changes in the chondroitin sulfate pathway in Dual-10 cells treated with butyrate. The current study is the first to report the effect of butyrate on glycosaminoglycan profiles.
Collapse
Affiliation(s)
- Payel Datta
- Department of Biology and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Avenue, Troy, NY, 12180, USA
| | | | | | | |
Collapse
|
25
|
Abstract
Syndecan-4, a ubiquitous cell surface proteoglycan, mediates numerous cellular processes through signaling pathways that affect cellular proliferation, migration, mechanotransduction and endocytosis. These effects are achieved through syndecan-4 functioning as both a co-receptor for the fibroblast growth factor receptors (FGFR1-FGFR4) and its ability to independently activate signaling pathways upon ligand binding. As an FGFR co-receptor, syndecan-4 strengthens the duration and intensity of downstream signaling upon ligand binding; this is particularly evident with regard to mitogen-activated protein kinase (MAPK) signaling. In contrast, syndecan-4 also functions as an independent receptor for heparin-binding growth factors, such as fibroblast growth factors (FGFs), vascular endothelial growth factors (VEGFs) and platelet-derived growth factors (PDGFs). These signaling cascades affect canonical signaling components, such as the mammalian target of rapamycin (mTOR), AKT1 and the Rho family of GTPases. In combination with the integrin family of proteins, syndecan-4 is also able to form physical connections between the extracellular matrix (ECM) and cytoskeletal signaling proteins, and it has a key role in regulation of integrin turnover. This unique versatility of the interactions of syndecan-4 is characterized in this Cell Science at a Glance article and illustrated in the accompanying poster.
Collapse
Affiliation(s)
- Arye Elfenbein
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06520, USA
| | | |
Collapse
|
26
|
Zheng X, Young Koh G, Jackson T. A continuous model of angiogenesis: Initiation, extension, and maturation of new blood vessels modulated by vascular endothelial growth factor, angiopoietins, platelet-derived growth factor-B, and pericytes. ACTA ACUST UNITED AC 2013. [DOI: 10.3934/dcdsb.2013.18.1109] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
27
|
Cattaruzza S, Ozerdem U, Denzel M, Ranscht B, Bulian P, Cavallaro U, Zanocco D, Colombatti A, Stallcup WB, Perris R. Multivalent proteoglycan modulation of FGF mitogenic responses in perivascular cells. Angiogenesis 2012; 16:309-27. [PMID: 23124902 DOI: 10.1007/s10456-012-9316-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Accepted: 10/12/2012] [Indexed: 01/10/2023]
Abstract
Sprouting of angiogenic perivascular cells is thought to be highly dependent upon autocrine and paracrine growth factor stimulation. Accordingly, we report that corneal angiogenesis induced by ectopic FGF implantation is strongly impaired in NG2/CSPG4 proteoglycan (PG) null mice known to harbour a putative deficit in pericyte proliferation/mobilization. Conversely, no significant differences were seen between wild type and knockout corneas when VEGF was used as an angiocrine factor. Perturbed responsiveness of NG2-deficient pericytes to paracrine and autocrine stimulation by several FGFs could be confirmed in cells isolated from NG2 null mice, while proliferation induced by other growth factors was equivalent in wild type and knockout cells. Identical results were obtained after siRNA-mediated knock-down of NG2 in human smooth muscle-like cell lines, as also demonstrated by the decreased levels of FGF receptor phosphorylation detected in these NG2 deprived cells. Binding assays with recombinant proteins and molecular interactions examined on live cells asserted that FGF-2 bound to NG2 in a glycosaminoglycan-independent, core protein-mediated manner and that the PG was alone capable of retaining FGF-2 on the cell membrane for subsequent receptor presentation. The use of dominant-negative mutant cells, engineered by combined transduction of NG2 deletion constructs and siRNA knock-down of the endogenous PG, allowed us to establish that the FGF co-receptor activity of NG2 is entirely mediated by its extracellular portion. In fact, forced overexpression of the NG2 ectodomain in human smooth muscle-like cells increased their FGF-2-induced mitosis and compensated for low levels of FGF receptor surface expression, in a manner equivalent to that produced by overexpression of the full-length NG2. Upon FGF binding, the cytoplasmic domain of NG2 is phosphorylated, but there is no evidence that this event elicits signal transductions that could bypass the FGFR-mediated ones. Pull-down experiments, protein-protein binding assays and flow cytometry FRET coherently revealed an elective ligand-independent association of NG2 with FGFR1 and FGFR3. The NG2 cooperation with these receptors was also corroborated functionally by the outcome of FGF-2 treatments of cells engineered to express diverse NG2/FGFR combinations. Comprehensively, the findings suggest that perivascular NG2 may serve as a dual modulator of the availability/accessibility of FGF at the cell membrane, as well as the resulting FGFR transducing activity.
Collapse
Affiliation(s)
- Sabrina Cattaruzza
- S.O.C. for Experimental Oncology 2, The National Cancer Institute Aviano, CRO-IRCCS, Via Pedemontana Occidentale 12, 33081, Aviano, PN, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Li L, Bennett SAL, Wang L. Role of E-cadherin and other cell adhesion molecules in survival and differentiation of human pluripotent stem cells. Cell Adh Migr 2012; 6:59-70. [PMID: 22647941 PMCID: PMC3364139 DOI: 10.4161/cam.19583] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The survival, proliferation, self-renewal and differentiation of human pluripotent stem cells (hPSCs, including human embryonic stem cells and human induced pluripotent stem cells) involve a number of processes that require cell-cell and cell-matrix interactions. The cell adhesion molecules (CAMs), a group of cell surface proteins play a pivotal role in mediating such interactions. Recent studies have provided insights into the essential roles and mechanisms of CAMs in the regulation of hPSC fate decisions. Here, we review the latest research progress in this field and focus on how E-cadherin and several other important CAMs including classic cadherins, Ig-superfamily CAMs, integrins and heparin sulfate proteoglycans control survival and differentiation of hPSCs.
Collapse
Affiliation(s)
- Li Li
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON Canada
| | | | | |
Collapse
|
29
|
|
30
|
Chen X, Xiao F, Wang Y, Fang J, Ding K. Structure-activity relationship study of WSS25 derivatives with anti-angiogenesis effects. Glycoconj J 2012; 29:389-98. [DOI: 10.1007/s10719-012-9424-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Revised: 06/18/2012] [Accepted: 06/25/2012] [Indexed: 12/12/2022]
|
31
|
Patel NS, Reisig KV, Clyne AM. A computational model of fibroblast growth factor-2 binding to endothelial cells under fluid flow. Ann Biomed Eng 2012; 41:154-71. [PMID: 22825797 DOI: 10.1007/s10439-012-0622-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 07/07/2012] [Indexed: 01/02/2023]
Abstract
Fibroblast growth factor-2 (FGF2) is an angiogenic growth factor that binds to cell surface receptors (FGFR) and heparan sulfate proteoglycans (HSPG), as well as HSPG in the basement membrane. FGF2 plays a critical role in angiogenesis, yet clinical FGF2 trials demonstrated limited success perhaps due to inadequate understanding of FGF2 binding in physiological conditions. We developed a computational model of FGF2 binding to isolated (HSPG or FGFR) or combined (HSPG and FGFR) binding sites under physiological fluid flow and predicted the effects of FGF2 concentration, binding site density, fluid flow rate, and delivery mode (continuous vs. bolus) on FGF2 complex formation. The isolated binding site models showed increased binding with FGF2 and binding site density. However, in the triad model, increasing FGF2 concentration decreased triads (FGF2-HSPG-FGFR) and increased FGF2-HSPG complexes. Fluid flow decreased time to equilibrium and dissociation in isolated binding site models, yet flow effect in the triad model depended on binding site density. Similarly, FGF2 capture and complex stability in bolus delivery depended on bolus size, flow rate, association and dissociation rate constants, as well as binding site density. This model shows the integrated effects of FGF2 binding stoichiometry, fluid flow, and delivery mode, and enhances our understanding of FGF2 complex formation under physiological conditions.
Collapse
Affiliation(s)
- Nisha S Patel
- Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
32
|
Haddad LE, Khzam LB, Hajjar F, Merhi Y, Sirois MG. Characterization of FGF receptor expression in human neutrophils and their contribution to chemotaxis. Am J Physiol Cell Physiol 2011; 301:C1036-45. [DOI: 10.1152/ajpcell.00215.2011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Several members of the fibroblast growth factor (FGF) family are potent endothelial cell (EC) mitogens and angiogenic factors, and their activities can be mediated by four tyrosine kinase receptors (FGFR1–4). In addition, FGFs can induce the release of inflammatory mediators by ECs and the expression of adhesion molecules at their surface, thereby favoring the recruitment and transvascular migration of inflammatory cells such as neutrophils. Neither the expression nor the biological activities that could be mediated by FGFRs have been investigated in human neutrophils. By biochemical and cytological analyses, we observed that purified circulating human neutrophils from healthy individuals expressed varying levels of FGFRs in their cytosol and at their cytoplasmic membrane. FGFR-2 was identified as the sole cell surface receptor, with FGFR-1 and -4 localizing in the cytosol and FGFR-3 being undetectable. We assessed the capacity of FGF-1 and FGF-2 to induce neutrophil chemotaxis in a modified Boyden microchamber and observed that they increase neutrophil transmigration at 10−10 and 10−9 M and by 1.77- and 2.34-fold, respectively, as compared with PBS-treated cells. Treatment with a selective anti-FGFR-2 antibody reduced FGF-1-mediated chemotaxis by 75% and abrogated the effect of FGF-2, while the blockade of FGFR-1 and -4 partially inhibited (15–40%) FGF-chemotactic activities. In summary, our data are the first to report the expression of FGF receptors in human neutrophils, with FGF-1 and FGF-2 promoting neutrophil chemotaxis mainly through FGFR-2 activation.
Collapse
Affiliation(s)
- Lydia E. Haddad
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Quebec, Canada; and
| | - Lara Bou Khzam
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Biomedical Sciences, Université de Montréal, Montreal, Quebec, Canada
| | - Fadi Hajjar
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Yahye Merhi
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Biomedical Sciences, Université de Montréal, Montreal, Quebec, Canada
| | - Martin G. Sirois
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Quebec, Canada; and
| |
Collapse
|
33
|
Re’em T, Cohen S. Microenvironment Design for Stem Cell Fate Determination. TISSUE ENGINEERING III: CELL - SURFACE INTERACTIONS FOR TISSUE CULTURE 2011; 126:227-62. [DOI: 10.1007/10_2011_118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
34
|
Zhao B, Zhang C, Forsten-Williams K, Zhang J, Fannon M. Endothelial cell capture of heparin-binding growth factors under flow. PLoS Comput Biol 2010; 6:e1000971. [PMID: 21060855 PMCID: PMC2965741 DOI: 10.1371/journal.pcbi.1000971] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2010] [Accepted: 09/23/2010] [Indexed: 11/18/2022] Open
Abstract
Circulation is an important delivery method for both natural and synthetic molecules, but microenvironment interactions, regulated by endothelial cells and critical to the molecule's fate, are difficult to interpret using traditional approaches. In this work, we analyzed and predicted growth factor capture under flow using computer modeling and a three-dimensional experimental approach that includes pertinent circulation characteristics such as pulsatile flow, competing binding interactions, and limited bioavailability. An understanding of the controlling features of this process was desired. The experimental module consisted of a bioreactor with synthetic endothelial-lined hollow fibers under flow. The physical design of the system was incorporated into the model parameters. The heparin-binding growth factor fibroblast growth factor-2 (FGF-2) was used for both the experiments and simulations. Our computational model was composed of three parts: (1) media flow equations, (2) mass transport equations and (3) cell surface reaction equations. The model is based on the flow and reactions within a single hollow fiber and was scaled linearly by the total number of fibers for comparison with experimental results. Our model predicted, and experiments confirmed, that removal of heparan sulfate (HS) from the system would result in a dramatic loss of binding by heparin-binding proteins, but not by proteins that do not bind heparin. The model further predicted a significant loss of bound protein at flow rates only slightly higher than average capillary flow rates, corroborated experimentally, suggesting that the probability of capture in a single pass at high flow rates is extremely low. Several other key parameters were investigated with the coupling between receptors and proteoglycans shown to have a critical impact on successful capture. The combined system offers opportunities to examine circulation capture in a straightforward quantitative manner that should prove advantageous for biologicals or drug delivery investigations.
Collapse
Affiliation(s)
- Bing Zhao
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, Kentucky, United States of America
| | - Changjiang Zhang
- Department of Computer Science, Laboratory for High Performance Computing and Computer Simulation, University of Kentucky, Lexington, Kentucky, United States of America
| | - Kimberly Forsten-Williams
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Jun Zhang
- Department of Computer Science, Laboratory for High Performance Computing and Computer Simulation, University of Kentucky, Lexington, Kentucky, United States of America
| | - Michael Fannon
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
35
|
Reisig K, Clyne AM. Fibroblast growth factor-2 binding to the endothelial basement membrane peaks at a physiologically relevant shear stress. Matrix Biol 2010; 29:586-93. [PMID: 20678572 DOI: 10.1016/j.matbio.2010.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Revised: 06/08/2010] [Accepted: 07/21/2010] [Indexed: 10/19/2022]
Abstract
Fibroblast growth factor-2 (FGF2) is produced and released by endothelial cells and binds to heparan sulfate proteoglycans in the endothelial basement membrane (BM), an important FGF2 storage reservoir. Experimental and computational models of FGF2 binding kinetics to both cells and BM under static conditions are well established in the literature but remain largely unexplored under flow. We now examine BM-FGF2 binding kinetics in fluid flow conditions. We hypothesized that FGF2 binding to the endothelial BM would decrease as fluid shear stress increased. To investigate this, BM-FGF2 equilibrium, associative, and dissociative bindings were measured at various shear stresses. Surprisingly, FGF2 binding increased up to a physiological arterial shear stress of 25 dynes/cm², after which it decreased to a level similar to the 1 dyne/cm² condition. Both BM-FGF2 dissociation and BM binding site availability increased with flow, while association remained constant. This suggests that force-dependent FGF2 equilibrium binding varies with shear stress due to a combination of an increase in binding site availability and FGF2 dissociation with flow. This improved understanding of BM-FGF2 binding with flow enriches current knowledge of FGF2 binding kinetics under physiologic conditions, which may contribute to improved growth factor therapy development.
Collapse
Affiliation(s)
- Karl Reisig
- Department of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | | |
Collapse
|
36
|
Peng SC, Wong DSH, Tung KC, Chen YY, Chao CC, Peng CH, Chuang YJ, Tang CY. Computational modeling with forward and reverse engineering links signaling network and genomic regulatory responses: NF-kappaB signaling-induced gene expression responses in inflammation. BMC Bioinformatics 2010; 11:308. [PMID: 20529327 PMCID: PMC2889938 DOI: 10.1186/1471-2105-11-308] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Accepted: 06/08/2010] [Indexed: 11/30/2022] Open
Abstract
Background Signal transduction is the major mechanism through which cells transmit external stimuli to evoke intracellular biochemical responses. Diverse cellular stimuli create a wide variety of transcription factor activities through signal transduction pathways, resulting in different gene expression patterns. Understanding the relationship between external stimuli and the corresponding cellular responses, as well as the subsequent effects on downstream genes, is a major challenge in systems biology. Thus, a systematic approach is needed to integrate experimental data and theoretical hypotheses to identify the physiological consequences of environmental stimuli. Results We proposed a systematic approach that combines forward and reverse engineering to link the signal transduction cascade with the gene responses. To demonstrate the feasibility of our strategy, we focused on linking the NF-κB signaling pathway with the inflammatory gene regulatory responses because NF-κB has long been recognized to play a crucial role in inflammation. We first utilized forward engineering (Hybrid Functional Petri Nets) to construct the NF-κB signaling pathway and reverse engineering (Network Components Analysis) to build a gene regulatory network (GRN). Then, we demonstrated that the corresponding IKK profiles can be identified in the GRN and are consistent with the experimental validation of the IKK kinase assay. We found that the time-lapse gene expression of several cytokines and chemokines (TNF-α, IL-1, IL-6, CXCL1, CXCL2 and CCL3) is concordant with the NF-κB activity profile, and these genes have stronger influence strength within the GRN. Such regulatory effects have highlighted the crucial roles of NF-κB signaling in the acute inflammatory response and enhance our understanding of the systemic inflammatory response syndrome. Conclusion We successfully identified and distinguished the corresponding signaling profiles among three microarray datasets with different stimuli strengths. In our model, the crucial genes of the NF-κB regulatory network were also identified to reflect the biological consequences of inflammation. With the experimental validation, our strategy is thus an effective solution to decipher cross-talk effects when attempting to integrate new kinetic parameters from other signal transduction pathways. The strategy also provides new insight for systems biology modeling to link any signal transduction pathways with the responses of downstream genes of interest.
Collapse
Affiliation(s)
- Shih Chi Peng
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Establishment of heparan sulphate deficient primary endothelial cells from EXT-1(flox/flox) mouse lungs and sprouting aortas. In Vitro Cell Dev Biol Anim 2010; 46:577-84. [PMID: 20383663 DOI: 10.1007/s11626-010-9313-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 03/11/2010] [Indexed: 10/19/2022]
Abstract
Angiogenesis is a hallmark of expanding tissue e.g. during embryogenesis and wound healing in physiology as well as in diseases such as cancer and atherosclerosis. Key steps of the angiogenic process involve growth factor-mediated stimulation of endothelial cell sprouting and tube formation. Heparan sulphate proteoglycans (HSPGs) have been implicated as important co-receptors of several pro-angiogenic proteins. The importance of HSPGs in physiology was underscored by the finding that knockout of the gene encoding HS polymerase, EXT-1, resulted in early embryonic lethality. Here, we describe the establishment of HS-deficient endothelial cells from sprouting aortas as well as from the lungs of EXT-1(flox/flox) mice. Recombination of the loxP-flanked EXT-1 locus by Cre-expressing adenovirus was demonstrated at the mRNA level. Moreover, depletion of HS polysaccharides was verified by flow cytometry and fluorescence microscopy methodology using phage display-derived anti-HS antibodies. In summary, we provide a genetic model to unravel the functional role of HSPGs specifically in primary endothelial cells during early steps of angiogenesis. Our studies are applicable to most loxP-based transgenic mouse strains, and may thus be of general importance in the angiogenesis field.
Collapse
|
38
|
Palamakumbura AH, Vora SR, Nugent MA, Kirsch KH, Sonenshein GE, Trackman PC. Lysyl oxidase propeptide inhibits prostate cancer cell growth by mechanisms that target FGF-2-cell binding and signaling. Oncogene 2009; 28:3390-400. [PMID: 19597471 PMCID: PMC2753565 DOI: 10.1038/onc.2009.203] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 06/05/2009] [Accepted: 06/14/2009] [Indexed: 11/08/2022]
Abstract
Enhanced RAS signaling and decreased androgen dependence of prostate cancer cells accompany poor clinical outcomes. Elevated autocrine fibroblast growth factors 2 (FGF-2) signaling promotes prostate cancer cell growth and survival. Expression of lysyl oxidase (LOX) inhibits RAS transforming activity. LOX is secreted as 50 kDa pro-LOX protein and then undergoes extracellular proteolytic processing to form approximately 30 kDa LOX enzyme and approximately 18 kDa propeptide (LOX-PP). We have previously shown that LOX-PP inhibits breast cancer cell transformation and tumor formation, but mechanisms of action of LOX-PP have not been fully elucidated. Here we report that LOX expression is reduced in prostate cancer cell lines and that recombinant LOX-PP protein inhibits serum-stimulated DNA synthesis and MEK/ERK and PI3K/AKT pathways in DU 145 and PC-3 androgen-independent cell lines. In DU 145 cells, treatment with a pharmacologic FGF-receptor inhibitor or a neutralizing anti-FGFR1 antibody mimicked LOX-PP inhibition of serum-stimulated DNA synthesis. FGF-2-stimulated DNA synthesis, ERK1/2, AKT and FRS2alpha activation were found all to be inhibited by LOX-PP in DU 145 cells. LOX-PP reduced specific binding of FGF-2 to DU 145 cells, suggesting that LOX-PP targets FGF signaling at the receptor. Interestingly, PC-3 cells did not respond to FGF-2, consistent with previous reports. We conclude that LOX-PP inhibits proliferation of DU 145 cells by interfering with FGFR(s) binding and signaling, and that LOX-PP has other mechanisms of action in PC-3 cells.
Collapse
Affiliation(s)
- Amitha H. Palamakumbura
- Department of Periodontology and Oral Biology, Division of Oral Biology, Boston University Goldman School of Dental Medicine, Boston, MA 02118
| | - Siddharth R. Vora
- Department of Periodontology and Oral Biology, Division of Oral Biology, Boston University Goldman School of Dental Medicine, Boston, MA 02118
| | - Matthew A. Nugent
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118
| | - Kathrin H. Kirsch
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118
| | - Gail E. Sonenshein
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118
| | - Philip C. Trackman
- Department of Periodontology and Oral Biology, Division of Oral Biology, Boston University Goldman School of Dental Medicine, Boston, MA 02118
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|
39
|
Tayalia P, Mooney DJ. Controlled growth factor delivery for tissue engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2009; 21:3269-3285. [PMID: 20882497 DOI: 10.1002/adma.200900241] [Citation(s) in RCA: 303] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Growth factors play a crucial role in information transfer between cells and their microenvironment in tissue engineering and regeneration. They initiate their action by binding to specific receptors on the surface of target cells and the chemical identity, concentration, duration, and context of these growth factors contain information that dictates cell fate. Hence, the importance of exogenous delivery of these molecules in tissue engineering is unsurprising, considering their importance for tissue regeneration. However, the short half-lives of growth factors, their relatively large size, slow tissue penetration, and their potential toxicity at high systemic levels, suggest that conventional routes of administration are unlikely to be effective. In this review, we provide an overview of the design criteria for growth factor delivery vehicles with respect to the growth factor itself and the microenvironment for delivery. We discuss various methodologies that could be adopted to achieve this localized delivery, and strategies using polymers as delivery vehicles in particular.
Collapse
|
40
|
Shen W, Zhang C, Fannon M, Forsten-Williams K, Zhang J. A computational model of FGF-2 binding and HSPG regulation under flow. IEEE Trans Biomed Eng 2009; 56:2147-55. [PMID: 19272950 PMCID: PMC2729807 DOI: 10.1109/tbme.2008.2002109] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A novel convection-diffusion-reaction model is developed to simulate fibroblast growth factor (FGF-2) binding to cell surface receptors (FGFRs) and heparan sulfate proteoglycans (HSPGs) under flow conditions within a cylindrical-shaped vessel or capillary. The model consists of a set of coupled nonlinear partial differential equations (PDEs) and a set of coupled nonlinear ordinary differential equations (ODEs). The time-dependent PDE system is discretized and solved by a second-order implicit Euler scheme using the finite volume method. The ODE system is solved by a stiff ODE solver VODE using backward differencing formulation (BDF). The transient solution of FGF-2, FGFR, HSPG, and their bound complexes for three different flow rates are computed and presented. Simulation results indicate that the model can predict growth factor transport and binding to receptors with/without the presence of heparan sulfate, as well as the effect of flow rate on growth factor-receptor binding. Our computational model may provide a useful means to investigate the impact of fluid flow on growth factor dynamics, and ultimately, signaling within the circulation.
Collapse
Affiliation(s)
- Wensheng Shen
- Department of Computational Science, SUNY Brockport, Brockport, NY, 14420 USA e-mail: (see http://www.cps.brockport.edu/~shen)
| | - Changjiang Zhang
- Department of Computer Science, University of Kentucky, Lexington, KY 40506 USA e-mail:
| | - Michael Fannon
- Department of Ophthamology and Visual Science, University of Kentucky, Lexington, KY 40506 USA e-mail:
| | - Kimberly Forsten-Williams
- Department of Chemical Engineering, Virginia Polytechnic Institute & State University, Blacksburg, VA 24601, USA e-mail:
| | - Jun Zhang
- Department of Computer Science, University of Kentucky, Lexington, KY 40506 USA e-mail: (see http://www.cs.uky.edu/~jzhang
| |
Collapse
|
41
|
Forsten-Williams K, Chu CL, Fannon M, Buczek-Thomas JA, Nugent MA. Control of growth factor networks by heparan sulfate proteoglycans. Ann Biomed Eng 2008; 36:2134-48. [PMID: 18839312 PMCID: PMC2606109 DOI: 10.1007/s10439-008-9575-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Accepted: 09/23/2008] [Indexed: 10/21/2022]
Abstract
Growth factor binding to transmembrane protein receptors is generally understood to initiate cell signaling. Receptor binding of heparin-binding growth factors (HB-GFs), such as fibroblast growth factor-2 (FGF-2), is regulated by interactions with heparan sulfate proteoglycans. While there is some specificity for binding to heparan sulfate, overlap in sites for different growth factors may allow for cross regulation. Here we demonstrate, using experiments and computer simulations, that the HB-GFs FGF-2 and heparin-binding EGF-like growth factor (HB-EGF) can cross regulate receptor binding of the other despite having unique receptors. The ability of HSPG to stabilize HB-GF receptor binding is critical for competing growth factors to modulate receptor binding with both enhanced and reduced binding possible depending on this stabilization process. HSPG density and affinity for HB-GF are also critical factors for HB-GF cross regulation. Simulations further reveal that HB-GF can regulate receptor binding of non-HB-GFs such as EGF even when the two proteins share no binding sites when other HB-GF are present within the network. Proliferation studies demonstrate potentiation of HB-EGF-induced growth by FGF-2 indicating that competition networks can alter biological response. Exogenous manipulation of cellular responses to growth factors in complex living systems will require understanding the HSPG-controlled network.
Collapse
Affiliation(s)
- Kimberly Forsten-Williams
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA.
| | | | | | | | | |
Collapse
|
42
|
Mac Gabhann F, Popel AS. Dimerization of VEGF receptors and implications for signal transduction: a computational study. Biophys Chem 2007; 128:125-39. [PMID: 17442480 PMCID: PMC2711879 DOI: 10.1016/j.bpc.2007.03.010] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2006] [Revised: 03/12/2007] [Accepted: 03/12/2007] [Indexed: 01/13/2023]
Abstract
Vascular endothelial growth factor (VEGF) is a potent cytokine involved in the induction of neovascularization. Secreted as a cysteine-linked dimer, it has two binding sites at opposite poles through which it may bind VEGF receptors (VEGFRs), receptor tyrosine kinases found on the surface of endothelial and other cells. The binding of a VEGF molecule to two VEGFR molecules induces transphosphorylation of the intracellular domains of the receptors, leading to signal transduction. The dominant mechanism of receptor dimerization is not clear: the receptors may be present in an inactive pre-dimerized form, VEGF binding first to one of the receptors, the second receptor then ideally located for dimerization; or VEGF may bind receptor monomers on the cell surface, which then diffuse and bind to available unligated receptor monomers to complete the activation. Both processes take place and one or other may dominate on different cell types. We demonstrate the impact of dimerization mechanism on the binding of VEGF to the cell surface and on the formation of active signaling receptor complexes. We describe two methods to determine which process dominates, based on binding and phosphorylation assays. The presence of two VEGF receptor populations, VEGFR1 and VEGFR2, can result in receptor heterodimer formation. Our simulations predict that heterodimers will comprise 10-50% of the active, signaling VEGF receptor complexes, and that heterodimers will form at the expense of homodimers of VEGFR1 when VEGFR2 populations are larger. These results have significant implications for VEGF signal transduction and interpretation of experimental studies. These results may be applicable to other ligand-receptor pairs, in particular PDGF.
Collapse
Affiliation(s)
- Feilim Mac Gabhann
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| | | |
Collapse
|
43
|
Abstract
Heparan sulphate proteoglycans reside on the plasma membrane of all animal cells studied so far and are a major component of extracellular matrices. Studies of model organisms and human diseases have demonstrated their importance in development and normal physiology. A recurrent theme is the electrostatic interaction of the heparan sulphate chains with protein ligands, which affects metabolism, transport, information transfer, support and regulation in all organ systems. The importance of these interactions is exemplified by phenotypic studies of mice and humans bearing mutations in the core proteins or the biosynthetic enzymes responsible for assembling the heparan sulphate chains.
Collapse
Affiliation(s)
- Joseph R Bishop
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California 92093, USA.
| | | | | |
Collapse
|
44
|
Fuster MM, Wang L, Castagnola J, Sikora L, Reddi K, Lee PHA, Radek KA, Schuksz M, Bishop JR, Gallo RL, Sriramarao P, Esko JD. Genetic alteration of endothelial heparan sulfate selectively inhibits tumor angiogenesis. J Cell Biol 2007; 177:539-49. [PMID: 17470635 PMCID: PMC2064806 DOI: 10.1083/jcb.200610086] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Accepted: 03/26/2007] [Indexed: 11/24/2022] Open
Abstract
To examine the role of endothelial heparan sulfate during angiogenesis, we generated mice bearing an endothelial-targeted deletion in the biosynthetic enzyme N-acetylglucosamine N-deacetylase/N-sulfotransferase 1 (Ndst1). Physiological angiogenesis during cutaneous wound repair was unaffected, as was growth and reproductive capacity of the mice. In contrast, pathological angiogenesis in experimental tumors was altered, resulting in smaller tumors and reduced microvascular density and branching. To simulate the angiogenic environment of the tumor, endothelial cells were isolated and propagated in vitro with proangiogenic growth factors. Binding of FGF-2 and VEGF(164) to cells and to purified heparan sulfate was dramatically reduced. Mutant endothelial cells also exhibited altered sprouting responses to FGF-2 and VEGF(164), reduced Erk phosphorylation, and an increase in apoptosis in branching assays. Corresponding changes in growth factor binding to tumor endothelium and apoptosis were also observed in vivo. These findings demonstrate a cell-autonomous effect of heparan sulfate on endothelial cell growth in the context of tumor angiogenesis.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Cell Line, Tumor
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/pathology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Fibroblast Growth Factor 2/pharmacology
- Heparitin Sulfate/metabolism
- Mice
- Mice, Mutant Strains
- Neoplasm Proteins/deficiency
- Neoplasm Proteins/metabolism
- Neoplasms, Experimental/enzymology
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Neovascularization, Pathologic/enzymology
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Organ Specificity/genetics
- Phosphorylation/drug effects
- Sulfotransferases/deficiency
- Sulfotransferases/metabolism
- Vascular Endothelial Growth Factor A/pharmacology
Collapse
Affiliation(s)
- Mark M Fuster
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Dupree MA, Pollack SR, Levine EM, Laurencin CT. Fibroblast growth factor 2 induced proliferation in osteoblasts and bone marrow stromal cells: a whole cell model. Biophys J 2006; 91:3097-112. [PMID: 16861274 PMCID: PMC1578487 DOI: 10.1529/biophysj.106.087098] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factor 2 (FGF2) can enhance the proliferative capacity of bone and bone marrow stromal cells; however, the mechanisms behind this effect are not well described. We present a whole-cell kinetic model relating receptor-mediated binding, internalization, and processing of FGF2 to osteoblastic proliferative response. Focusing on one of the potential signaling complex stoichiometries, we utilized experimentally measured and modeled estimated rate constants to predict in vitro proliferation and distinguish between potential binding orders. We found that piecewise assemblage of a ternary signaling complex may occur in several ways depending on the local binding environment. Using experimental data of endocytosed FGF2 as a constraint, we have also shown evidence of potential multistep processes involved in heparan-sulfate proteoglycans-bound FGF2 release, internalization, and fragment formation in conjunction with the normal metabolism of the proteoglycan.
Collapse
Affiliation(s)
- Melissa A Dupree
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | |
Collapse
|
46
|
Muir C, Chung LWK, Carson DD, Farach-Carson MC. Hypoxia increases VEGF-A production by prostate cancer and bone marrow stromal cells and initiates paracrine activation of bone marrow endothelial cells. Clin Exp Metastasis 2006; 23:75-86. [PMID: 16826426 DOI: 10.1007/s10585-006-9021-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Accepted: 05/14/2006] [Indexed: 11/29/2022]
Abstract
Hypoxia develops at sites of rapid cancer growth near sites of poorly organized vasculature. Heparin binding growth factors (HBGFs) support neoangiogenesis of tumors. We examined the effect of culturing bone-targeted, metastatic C4-2B prostate cancer cells and bone stromal derived HS27a cells under hypoxic conditions on expression of vascular endothelial growth factor (VEGF) family members. A sealed chamber infused with 1% (hypoxic) or 20% (normoxic) O(2) was used. Both cell lines produced VEGF-A in normoxia, but little or no HB-EGF, another HBGF. HS27a cells produced low levels of FGF-2 and HGF, but little or none was secreted by C4-2B cells. Levels of VEGF-A in conditioned medium (CM) from both cell lines doubled when cultured in hypoxia. Similar changes in VEGF-A mRNA levels were seen. Receptor expression was unchanged by hypoxia. Changes in VEGF-A expression during hypoxia were preceded by nuclear accumulation of hypoxia inducible factor-1alpha (HIF-1alpha). Bone marrow endothelial (BME) cells express high levels of VEGFR2/flk-1, and are targets of VEGF-A induced neovascularization. BME cells proliferated in response to treatment with HS27a CM, but not C4-2B CM. BME cells formed tube-like angiogenic structures on growth factor reduced Matrigel in response to CM from HS27a or C4-2B cells. This response was greater when CM was produced under hypoxia, and was reduced by VEGF-A or FGF-2 neutralizing antibodies. We conclude that hypoxia triggers a physiologically relevant increase in VEGF-A by prostate cancer and bone marrow stromal cells which involves a paracrine loop that recruits and activates BME to support tumor neovascularization-related processes.
Collapse
Affiliation(s)
- Caroline Muir
- Department of Biological Sciences, University of Delaware, Room 326 Wolf Hall, Newark, DE 19716, USA
| | | | | | | |
Collapse
|
47
|
Krishna D, Le Doux JM. Murine leukemia virus particles activate Rac1 in HeLa cells. Biochem Biophys Res Commun 2006; 345:1184-93. [PMID: 16716260 DOI: 10.1016/j.bbrc.2006.05.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2006] [Accepted: 05/05/2006] [Indexed: 12/19/2022]
Abstract
A number of viruses, when they bind to cells, activate intracellular signals that facilitate post-binding steps of infection. To determine if retroviruses activate intracellular signaling, we transduced HeLa cells with amphotropic retroviruses produced by TelCeB6 cells and examined cell lysates for activated Rac1. We found that retroviruses activate Rac1. Rac1 activation was blocked when cells were depleted of cholesterol, cultured in suspension, or incubated with an anti-beta(1) integrin antibody, and when viruses were treated with heparinase III. Retrovirus activation of Rac1 did not require the amphotropic envelope protein. Gene transfer was reduced 2.4-fold when viruses were treated with heparinase III, but did not change when cells were transduced in the presence of function-blocking anti-beta(1) integrin antibodies. The implications of these findings with respect to retrovirus-cell interactions are discussed.
Collapse
Affiliation(s)
- Delfi Krishna
- The School of Chemical and Biomolecular Engineering, Atlanta, GA 30332-0535, USA
| | | |
Collapse
|
48
|
Leadbeater WE, Gonzalez AM, Logaras N, Berry M, Turnbull JE, Logan A. Intracellular trafficking in neurones and glia of fibroblast growth factor-2, fibroblast growth factor receptor 1 and heparan sulphate proteoglycans in the injured adult rat cerebral cortex. J Neurochem 2006; 96:1189-200. [PMID: 16417571 DOI: 10.1111/j.1471-4159.2005.03632.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The potent gliogenic and neurotrophic fibroblast growth factor (FGF)-2 signals through a receptor complex comprising high-affinity FGF receptor (FGFR)1 with heparan sulphate proteoglycans (HSPGs) as co-receptors. We examined the intracellular dynamics of FGF-2, FGFR1 and the HSPGs syndecan-2 and -3, glypican-1 and -2, and perlecan in neurones and glia in and around adult rat cerebral wounds. In the intact cerebral cortex, FGF-2 and FGFR1 mRNA and protein were constitutively expressed in astrocytes and neurones respectively. FGF-2 protein was localized exclusively to astrocyte nuclei. After injury, expression of FGF-2 mRNA was up-regulated only in astrocytes, whereas FGFR1 mRNA expression was increased in both glia and neurones, a disparity indicating that FGF-2 may act as a paracrine and autocrine factor for neurones and glia respectively. FGF-2 protein localized to both cytoplasm and nuclei of injury-responsive neurones and glia. There was weak or no staining of HSPGs in the normal cerebral neuropil and glia nuclei, with a few immunopositive neurones. Specific HSPGs responded to injury by differentially co-localizing with trafficked intracellular FGF-2 and FGFR1. The spatiotemporal dynamics of FGF-2-FGFR1-HSPG complex formation implies a role for individual HSPGs in regulating FGF-2 storage, nuclear trafficking and cell-specific injury responses in CNS wounds.
Collapse
Affiliation(s)
- W E Leadbeater
- Molecular Neuroscience Group, Division of Medical Sciences, University of Birmingham, Birmingham, UK.
| | | | | | | | | | | |
Collapse
|
49
|
Ray PE, Tassi E, Liu XH, Wellstein A. Role of fibroblast growth factor-binding protein in the pathogenesis of HIV-associated hemolytic uremic syndrome. Am J Physiol Regul Integr Comp Physiol 2006; 290:R105-13. [PMID: 16352855 DOI: 10.1152/ajpregu.00492.2005] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A characteristic finding of childhood HIV-associated hemolytic uremic syndrome (HIV-HUS) is the presence of endothelial injury and microcystic tubular dilation, leading to a rapid progression of the renal disease. We have previously shown that a secreted fibroblast growth factor-binding protein (FGF-BP) is upregulated in kidneys from children affected with HIV-HUS and HIV nephropathy. Here, we sought to determine the potential role of FGF-BP in the pathogenesis of HIV-HUS. By immunohistochemical and in situ hybridization studies, we observed FGF-BP protein and mRNA upregulation in regenerating renal tubular epithelial cells from kidneys of HIV-Tg26mice with late-stage renal disease, that is, associated with the development of microcystic tubular dilatation and accumulation of FGF-2. Moreover, FGF-BP increased the FGF-2-dependent growth and survival of cultured primary human renal glomerular endothelial cells and enhanced FGF-2-induced MAPK/ERK2 activation, as well as the proliferation of immortalized GM7373 endothelial cells. We propose that HIV-Tg26mice are a clinically relevant model system to study the role of FGF-BP in the pathogenesis of HIV-associated renal diseases. Furthermore, the upregulation of FGF-BP by regenerating renal tubular epithelial cells may provide a mechanism by which the regenerative and angiogenic activity of FGF-2 in renal capillaries can be modulated in children with HIV-HUS and other renal disease.
Collapse
Affiliation(s)
- Patricio E Ray
- Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Rd., Washington DC 20057, USA
| | | | | | | |
Collapse
|
50
|
Chu CL, Goerges AL, Nugent MA. Identification of common and specific growth factor binding sites in heparan sulfate proteoglycans. Biochemistry 2005; 44:12203-13. [PMID: 16142919 DOI: 10.1021/bi050241p] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The structural complexity within heparan sulfate has suggested that it contains multiple protein-specific binding sites. To evaluate the selectivity of growth factor binding to heparan sulfate, we conducted a detailed study of the intercompetition of fibroblast growth factor-2 (FGF-2) and heparin-binding epidermal growth factor-like growth factor (HB-EGF) binding to heparan sulfate (HS) on bovine aortic smooth muscle cells. Radioligand binding assays were conducted, and an analytical method was developed for determining the apparent binding constants and numbers of specific and shared binding sites within HS. These studies revealed the presence of two general classes of HS-binding sites for FGF-2 and HB-EGF. The major class (approximately 10(6) sites per cell) was able to bind to either growth factor with relatively low affinity (K(d) = 12 and 44 nM for FGF-2 and HB-EGF, respectively) and was termed "common" binding sites. However, both FGF-2 and HB-EGF also showed specific high affinity (0.6 and 6.1 nM for FGF-2 and HB-EGF, respectively) binding to a minor subset (118,000 and 28,000 sites per cell for FGF-2 and HB-EGF, respectively) of "unique" binding sites, which were unable to bind the other growth factor. These studies indicate that growth factor binding to HS involves multiple binding sites of variable affinity, density, and selectivity. The approach outlined in this study could be applied to aid in the evaluation of the relative biological roles of these selective and nonselective growth factor binding domains within HS.
Collapse
Affiliation(s)
- Chia Lin Chu
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | |
Collapse
|