1
|
Everts PA, Lana JF, Alexander RW, Dallo I, Kon E, Ambach MA, van Zundert A, Podesta L. Profound Properties of Protein-Rich, Platelet-Rich Plasma Matrices as Novel, Multi-Purpose Biological Platforms in Tissue Repair, Regeneration, and Wound Healing. Int J Mol Sci 2024; 25:7914. [PMID: 39063156 PMCID: PMC11277244 DOI: 10.3390/ijms25147914] [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: 05/26/2024] [Revised: 07/07/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Autologous platelet-rich plasma (PRP) preparations are prepared at the point of care. Centrifugation cellular density separation sequesters a fresh unit of blood into three main fractions: a platelet-poor plasma (PPP) fraction, a stratum rich in platelets (platelet concentrate), and variable leukocyte bioformulation and erythrocyte fractions. The employment of autologous platelet concentrates facilitates the biological potential to accelerate and support numerous cellular activities that can lead to tissue repair, tissue regeneration, wound healing, and, ultimately, functional and structural repair. Normally, after PRP preparation, the PPP fraction is discarded. One of the less well-known but equally important features of PPP is that particular growth factors (GFs) are not abundantly present in PRP, as they reside outside of the platelet alpha granules. Precisely, insulin-like growth factor-1 (IGF-1) and hepatocyte growth factor (HGF) are mainly present in the PPP fraction. In addition to their roles as angiogenesis activators, these plasma-based GFs are also known to inhibit inflammation and fibrosis, and they promote keratinocyte migration and support tissue repair and wound healing. Additionally, PPP is known for the presence of exosomes and other macrovesicles, exerting cell-cell communication and cell signaling. Newly developed ultrafiltration technologies incorporate PPP processing methods by eliminating, in a fast and efficient manner, plasma water, cytokines, molecules, and plasma proteins with a molecular mass (weight) less than the pore size of the fibers. Consequently, a viable and viscous protein concentrate of functional total proteins, like fibrinogen, albumin, and alpha-2-macroglobulin is created. Consolidating a small volume of high platelet concentrate with a small volume of highly concentrated protein-rich PPP creates a protein-rich, platelet-rich plasma (PR-PRP) biological preparation. After the activation of proteins, mainly fibrinogen, the PR-PRP matrix retains and facilitates interactions between invading resident cells, like macrophages, fibroblast, and mesenchymal stem cells (MSCs), as well as the embedded concentrated PRP cells and molecules. The administered PR-PRP biologic will ultimately undergo fibrinolysis, leading to a sustained release of concentrated cells and molecules that have been retained in the PR-PRP matrix until the matrix is dissolved. We will discuss the unique biological and tissue reparative and regenerative properties of the PR-PRP matrix.
Collapse
Affiliation(s)
- Peter A. Everts
- Gulf Coast Biologics, A Non-Profit Organization, Fort Myers, FL 33916, USA
- OrthoRegen Group, Max-Planck University, Indaiatuba 13334-170, SP, Brazil;
| | - José Fábio Lana
- OrthoRegen Group, Max-Planck University, Indaiatuba 13334-170, SP, Brazil;
| | - Robert W. Alexander
- Regenevita Biocellular Aesthetic & Reconstructive Surgery, Cranio-Maxillofacial Surgery, Regenerative and Wound Healing, Hamilton, MT 59840, USA;
- Department of Surgery & Maxillofacial Surgery, School of Medicine & Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Ignacio Dallo
- Unit of Biological Therapies and MSK Interventionism, Department of Orthopaedic Surgery and Sports Medicine, Sport Me Medical Center, 41013 Seville, Spain;
| | - Elizaveta Kon
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Mary A. Ambach
- BioEvolve, San Diego Orthobiologics and Sports Center, San Diego, CA 92024, USA
| | - André van Zundert
- Department of Anaesthesia and Perioperative Medicine, Royal Brisbane and Women’s Hospital, Brisbane and The University of Queensland, Brisbane 4072, Australia;
| | - Luga Podesta
- Bluetail Medical Group & Podesta Orthopedic Sports Medicine, Naples, FL 34109, USA;
- Physical Medicine & Rehabilitation Orlando College of Osteopathic Medicine, Orlando, FL 32806, USA
| |
Collapse
|
2
|
Yakovlev S, Tjandra N, Strickland DK, Medved L. Identification of Neural (N)-Cadherin as a Novel Endothelial Cell Receptor for Fibrin and Localization of the Complementary Binding Sites. Biochemistry 2024; 63:202-211. [PMID: 38156948 PMCID: PMC10848343 DOI: 10.1021/acs.biochem.3c00475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Based on the high structural homology between vascular endothelial (VE)-cadherin and neural (N)-cadherin, we hypothesized that fibrin, which is known to interact with VE-cadherin and promote angiogenesis through this interaction, may also interact with N-cadherin. To test this hypothesis, we prepared fibrin and its plasmin-produced and recombinant fragments covering practically all parts of the fibrin molecule. We also prepared the soluble extracellular portion of N-cadherin (sN-cadherin), which includes all five extracellular N-cadherin domains, and studied its interaction with fibrinogen, fibrin, and the aforementioned fibrin fragments using two independent methods, ELISA and SPR. The experiments confirmed our hypothesis, revealing that fibrin interacts with sN-cadherin with high affinity. Furthermore, the experiments localized the N-cadherin binding site within the fibrin βN-domains. Notably, the recombinant dimeric (β15-66)2 fragment, corresponding to these domains and mimicking their dimeric arrangement in fibrin, preserved the N-cadherin-binding properties of fibrin. To localize the fibrin binding site within N-cadherin, we performed ELISA and SPR experiments with (β15-66)2 and recombinant N-cadherin fragments representing its individual extracellular domains and combinations thereof. The results obtained indicate that the interaction of fibrin with N-cadherin occurs through the third and fifth extracellular domains of the latter. This is in contrast to our previous study, which revealed that fibrin interacts only with the third extracellular domain of VE-cadherin. In conclusion, our study identified N-cadherin as a novel receptor for fibrin and localized complementary binding sites within both fibrin and N-cadherin. The pathophysiological role of this interaction remains to be established.
Collapse
Affiliation(s)
- Sergiy Yakovlev
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Nico Tjandra
- Laboratory of Structural Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Dudley K. Strickland
- Center for Vascular and Inflammatory Diseases and Departments of Surgery and Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Leonid Medved
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| |
Collapse
|
3
|
Angelidakis E, Chen S, Zhang S, Wan Z, Kamm RD, Shelton SE. Impact of Fibrinogen, Fibrin Thrombi, and Thrombin on Cancer Cell Extravasation Using In Vitro Microvascular Networks. Adv Healthc Mater 2023; 12:e2202984. [PMID: 37119127 PMCID: PMC10524192 DOI: 10.1002/adhm.202202984] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/25/2023] [Indexed: 04/30/2023]
Abstract
A bidirectional association exists between metastatic dissemination and the hypercoagulable state associated with many types of cancer. As such, clinical studies have provided evidence that markers associated with elevated levels of coagulation and fibrinolysis correlate with decreased patient survival. However, elucidating the mechanisms underpinning the effects of different components of the coagulation system on metastasis formation is challenging both in animal models and 2D models lacking the complex cellular interactions necessary to model both thrombosis and metastasis. Here, an in vitro, 3D, microvascular model for observing the formation of fibrin thrombi is described, which is in turn used to study how different aspects of the hypercoagulable state associated with cancer affect the endothelium. Using this platform, cancer cells expressing ICAM-1 are shown to form a fibrinogen-dependent bridge and transmigrate through the endothelium more effectively. Cancer cells are also demonstrated to interact with fibrin thrombi, using them to adhere, spread, and enhance their extravasation efficiency. Finally, thrombin is also shown to enhance cancer cell extravasation. This system presents a physiologically relevant model of fibrin clot formation in the human microvasculature, enabling in-depth investigation of the cellular interactions between cancer cells and the coagulation system affecting cancer cell extravasation.
Collapse
Affiliation(s)
- Emmanouil Angelidakis
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sophia Chen
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Shun Zhang
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Zhengpeng Wan
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Roger D. Kamm
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sarah E. Shelton
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Medical OncologyDana Farber Cancer InstituteBostonMA02215USA
| |
Collapse
|
4
|
Díaz-Flores L, Gutiérrez R, Pino García M, González-Gómez M, Díaz-Flores L, Carrasco JL, Madrid JF, Álvarez-Argüelles H. Intussusceptive angiogenesis facilitated by microthrombosis has an important example in angiolipoma. An ultrastructural and immunohistochemical study. Histol Histopathol 2023; 38:29-46. [PMID: 35775452 DOI: 10.14670/hh-18-488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The microvasculature of angiolipoma frequently presents thrombi. Our objectives are to assess whether intussusceptive angiogenesis (IA) participates in vasculature formation in non-infiltrating angiolipoma and, if so, to explore how thrombi are involved in the IA process. For this purpose, we studied angiolipoma specimens (n: 52), using immunohistochemistry, and confocal and electron microscopy. The results showed the presence of folds and pillars, hallmarks of IA, dividing the vessel lumen. Folds showed a cover formed by reoriented endothelial cells from the vessel wall, or from newly formed folds, and a core initially formed by thrombus fragments (clot components as transitional core), which was replaced by extracellular matrix and invaginating pericytes establishing numerous peg-and-socket junctions with endothelial cells (mature core). A condensed plasmatic electron-dense material surrounded and connected folds and pillars with each other and with the vascular wall, which suggests a clot role in fold/pillar arrangement. In conclusion, we contribute to IA participation in capillary network formation in angiolipoma and the immunohistochemical and ultrastructural events by which microthrombosis facilitates IA. Therefore, in addition to the histogenesis of angiolipoma, we provide an easily obtainable substrate for future studies on clot component action in IA, of clinical and therapeutic interest.
Collapse
Affiliation(s)
- Lucio Díaz-Flores
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, Tenerife, Spain.
| | - Ricardo Gutiérrez
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | - Maria Pino García
- Department of Pathology, Eurofins Megalab-Hospiten Hospitals, Tenerife, Spain
| | - Miriam González-Gómez
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomédicas de Canarias, University of La Laguna, Tenerife, Spain
| | - Lucio Díaz-Flores
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | - Jose Luis Carrasco
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | - Juan Francisco Madrid
- Department of Cell Biology and Histology, School of Medicine, Campus of International Excellence "Campus Mare Nostrum", IMIB-Arrixaca, University of Murcia, Murcia, Spain
| | - Hugo Álvarez-Argüelles
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| |
Collapse
|
5
|
Yakovlev S, Strickland DK, Medved L. Current View on the Molecular Mechanisms Underlying Fibrin(ogen)-Dependent Inflammation. Thromb Haemost 2022; 122:1858-1868. [PMID: 35896433 PMCID: PMC10680782 DOI: 10.1055/a-1910-4538] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Numerous studies have revealed the involvement of fibrinogen in the inflammatory response. To explain the molecular mechanisms underlying fibrinogen-dependent inflammation, two bridging mechanisms have been proposed in which fibrin(ogen) bridges leukocytes to endothelial cells. The first mechanism suggests that bridging occurs via the interaction of fibrinogen with the leukocyte receptor Mac-1 and the endothelial receptor ICAM-1 (intercellular adhesion molecule-1), which promotes leukocyte transmigration and enhances inflammation. The second mechanism includes bridging of leukocytes to the endothelium by fibrin degradation product E1 fragment through its interaction with leukocyte receptor CD11c and endothelial VE-cadherin to promote leukocyte transmigration. The role of E1 in promoting inflammation is inhibited by the fibrin-derived β15-42 fragment, and this has been suggested to result from its ability to compete for the E1-VE-cadherin interaction and to trigger signaling pathways through the src kinase Fyn. Our recent study revealed that the β15-42 fragment is ineffective in inhibiting the E1- or fibrin-VE-cadherin interaction, leaving the proposed signaling mechanism as the only viable explanation for the inhibitory function of β15-42. We have discovered that fibrin interacts with the very-low-density lipoprotein (VLDL) receptor, and this interaction triggers a signaling pathway that promotes leukocyte transmigration through inhibition of the src kinase Fyn. This pathway is inhibited by another pathway induced by the interaction of β15-42 with a putative endothelial receptor. In this review, we briefly describe the previously proposed molecular mechanisms underlying fibrin-dependent inflammation and their advantages/disadvantages and summarize our recent studies of the novel VLDL receptor-dependent pathway of leukocyte transmigration which plays an important role in fibrin-dependent inflammation.
Collapse
Affiliation(s)
- Sergiy Yakovlev
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Dudley K. Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Leonid Medved
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
6
|
Yakovlev S, Medved L. Dual functions of the fibrin βN-domains in the VLDL receptor-dependent pathway of transendothelial migration of leukocytes. Thromb Res 2022; 214:1-7. [PMID: 35421681 PMCID: PMC10680947 DOI: 10.1016/j.thromres.2022.04.002] [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: 02/17/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 11/16/2022]
Abstract
Our previous studies revealed that fibrin interacts with the VLDL receptor (VLDLR) through a pair of its βN-domains and this interaction promotes transendothelial migration of leukocytes and, thereby, inflammation. In agreement, the NDSK-II fragment representing the central part of the fibrin molecule and containing these domains stimulates leukocyte transmigration. However, the recombinant (β15-66)2 fragment corresponding to a pair of the βN-domains inhibits NDSK-II-stimulated leukocyte transmigration. To explain this paradox, we hypothesized that fibrin βN-domains have dual function in fibrin-dependent inflammation, namely, their C-terminal regions containing the VLDLR-binding sites promote leukocyte transmigration while their N-terminal regions are responsible for inhibition of this process. To test this hypothesis and to further clarify the molecular mechanisms underlying fibrin-induced VLDLR-dependent pathway of leukocyte transmigration and its inhibition, we prepared the dimeric (β15-44)2 and (β40-66)2 fragments corresponding to the N- and C-terminal regions of the βN-domains and studied their effect on endothelial permeability and transendothelial migration of leukocytes. The results obtained revealed that (β40-66)2 bound to the VLDLR with high affinity and promoted endothelial permeability and leukocyte transmigration while (β15-44)2 did not interact with this receptor and had no effect on leukocyte transmigration, in agreement with our hypothesis. We also found that the first three N-terminal residues of the βN-domains play a critical role in the inhibitory properties of these domains. Further, the inhibitory properties of the βN-domains were expressed only upon their isolation from the fibrin molecule. The question of whether their inhibitory function may play a role in fibrin remains to be addressed.
Collapse
Affiliation(s)
- Sergiy Yakovlev
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Leonid Medved
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
7
|
Liu K, Wiendels M, Yuan H, Ruan C, Kouwer PH. Cell-matrix reciprocity in 3D culture models with nonlinear elasticity. Bioact Mater 2022; 9:316-331. [PMID: 34820573 PMCID: PMC8586441 DOI: 10.1016/j.bioactmat.2021.08.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/24/2021] [Accepted: 08/03/2021] [Indexed: 01/17/2023] Open
Abstract
Three-dimensional (3D) matrix models using hydrogels are powerful tools to understand and predict cell behavior. The interactions between the cell and its matrix, however is highly complex: the matrix has a profound effect on basic cell functions but simultaneously, cells are able to actively manipulate the matrix properties. This (mechano)reciprocity between cells and the extracellular matrix (ECM) is central in regulating tissue functions and it is fundamentally important to broadly consider the biomechanical properties of the in vivo ECM when designing in vitro matrix models. This manuscript discusses two commonly used biopolymer networks, i.e. collagen and fibrin gels, and one synthetic polymer network, polyisocyanide gel (PIC), which all possess the characteristic nonlinear mechanics in the biological stress regime. We start from the structure of the materials, then address the uses, advantages, and limitations of each material, to provide a guideline for tissue engineers and biophysicists in utilizing current materials and also designing new materials for 3D cell culture purposes.
Collapse
Affiliation(s)
- Kaizheng Liu
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Radboud University, Institute for Molecules and Materials, Heyendaalseweg 135, 6525 AJ, Nijmegen, the Netherlands
| | - Maury Wiendels
- Radboud University, Institute for Molecules and Materials, Heyendaalseweg 135, 6525 AJ, Nijmegen, the Netherlands
| | - Hongbo Yuan
- Institute of Biophysics, Hebei University of Technology, Tianjin, 300401, PR China
- Molecular Imaging and Photonics, Chemistry Department, KU Leuven, Celestijnenlaan 200F, 3001, Heverlee, Belgium
| | - Changshun Ruan
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Paul H.J. Kouwer
- Radboud University, Institute for Molecules and Materials, Heyendaalseweg 135, 6525 AJ, Nijmegen, the Netherlands
| |
Collapse
|
8
|
Harki O, Faury G, Vilgrain I, Pépin JL, Briançon-Marjollet A. Reply: Soluble VE-cadherin: not just a marker of endothelial permeability. Eur Respir J 2021; 58:13993003.02629-2021. [PMID: 34711535 DOI: 10.1183/13993003.02629-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/11/2021] [Indexed: 02/03/2023]
Affiliation(s)
- Olfa Harki
- Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, HP2, Grenoble, France
| | - Gilles Faury
- Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, HP2, Grenoble, France
| | | | - Jean-Louis Pépin
- Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, HP2, Grenoble, France
| | | |
Collapse
|
9
|
Gruschus JM, Yakovlev S, Banerjee K, Medved L, Tjandra N. Structural Basis for the Interaction of Fibrin with the Very Low-Density Lipoprotein Receptor Revealed by NMR and Site-Directed Mutagenesis. Biochemistry 2021; 60:2537-2548. [PMID: 34351135 PMCID: PMC10658778 DOI: 10.1021/acs.biochem.1c00378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Interaction of fibrin with the very low-density lipoprotein receptor (VLDLR) promotes transendothelial migration of leukocytes and thereby inflammation. To establish the structural basis for this interaction, we have previously localized the VLDLR-binding site to fibrin βN-domains including fibrin β chain sequence 15-64 and determined the NMR solution structure of the VLDLR(2-4) fragment containing fibrin-binding CR domains 2-4 of VLDLR. In this study, we identified amino acid residues in VLDLR and the βN-domains that are involved in the interaction using NMR and site-directed mutagenesis. The results obtained revealed that Lys47 and Lys53 of the second and third positively charged clusters of the βN-domain, respectively, interact with Trp20 and Asp25 of the CR2 domain and Trp63 and Glu68 of the CR3 domain, respectively. This finding indicates that Lys residues of the βN-domain interact with the Lys-binding site of the CR domains in a manner proposed earlier for the interaction of other members of the LDL receptor family with their ligands. In addition, Gly15 of the βN-domain and its first positively charged cluster contribute to the high-affinity interaction with VLDLR. Molecular modeling based on the results obtained and analysis of the previously published structures of such domains complexed with RAP and HRV2 allowed us to propose a model of interaction of fibrin βN-domains with the fibrin-binding CR domains of the VLDL receptor.
Collapse
Affiliation(s)
- James M. Gruschus
- Laboratory of Structural Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Sergiy Yakovlev
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Koyeli Banerjee
- Laboratory of Structural Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Leonid Medved
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Nico Tjandra
- Laboratory of Structural Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
10
|
Joyce K, Fabra GT, Bozkurt Y, Pandit A. Bioactive potential of natural biomaterials: identification, retention and assessment of biological properties. Signal Transduct Target Ther 2021; 6:122. [PMID: 33737507 PMCID: PMC7973744 DOI: 10.1038/s41392-021-00512-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/29/2020] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
Biomaterials have had an increasingly important role in recent decades, in biomedical device design and the development of tissue engineering solutions for cell delivery, drug delivery, device integration, tissue replacement, and more. There is an increasing trend in tissue engineering to use natural substrates, such as macromolecules native to plants and animals to improve the biocompatibility and biodegradability of delivered materials. At the same time, these materials have favourable mechanical properties and often considered to be biologically inert. More importantly, these macromolecules possess innate functions and properties due to their unique chemical composition and structure, which increase their bioactivity and therapeutic potential in a wide range of applications. While much focus has been on integrating these materials into these devices via a spectrum of cross-linking mechanisms, little attention is drawn to residual bioactivity that is often hampered during isolation, purification, and production processes. Herein, we discuss methods of initial material characterisation to determine innate bioactivity, means of material processing including cross-linking, decellularisation, and purification techniques and finally, a biological assessment of retained bioactivity of a final product. This review aims to address considerations for biomaterials design from natural polymers, through the optimisation and preservation of bioactive components that maximise the inherent bioactive potency of the substrate to promote tissue regeneration.
Collapse
Affiliation(s)
- Kieran Joyce
- School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Georgina Targa Fabra
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Yagmur Bozkurt
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland.
| |
Collapse
|
11
|
Li L, Liu Q, Shang T, Song W, Xu D, Allen TD, Wang X, Jeong J, Lobe CG, Liu J. Aberrant Activation of Notch1 Signaling in Glomerular Endothelium Induces Albuminuria. Circ Res 2021; 128:602-618. [PMID: 33435713 DOI: 10.1161/circresaha.120.316970] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Glomerular capillaries are lined with a highly specialized fenestrated endothelium and contribute to the glomerular filtration barrier. The Notch signaling pathway is involved in regulation of glomerular filtration barrier, but its role in glomerular endothelium has not been investigated due to the embryonic lethality of animal models with genetic modification of Notch pathway components in the endothelium. OBJECTIVE To determine the effects of aberrant activation of the Notch signaling in glomerular endothelium and the underlying molecular mechanisms. METHODS AND RESULTS We established the ZEG-NICD1 (notch1 intracellular domain)/Tie2-tTA/Tet-O-Cre transgenic mouse model to constitutively activate Notch1 signaling in endothelial cells of adult mice. The triple transgenic mice developed severe albuminuria with significantly decreased VE-cadherin (vascular endothelial cadherin) expression in the glomerular endothelium. In vitro studies showed that either NICD1 (Notch1 intracellular domain) lentiviral infection or treatment with Notch ligand DLL4 (delta-like ligand 4) markedly reduced VE-cadherin expression and increased monolayer permeability of human renal glomerular endothelial cells. In addition, Notch1 activation or gene knockdown of VE-cadherin reduced the glomerular endothelial glycocalyx. Further investigation demonstrated that activated Notch1 suppression of VE-cadherin was through the transcription factors SNAI1 (snail family transcriptional repressor 1) and ERG (Ets related gene), which bind to the -373 E-box and the -134/-118 ETS (E26 transformation-specific) element of the VE-cadherin promoter, respectively. CONCLUSIONS Our results reveal novel regulatory mechanisms whereby endothelial Notch1 signaling dictates the level of VE-cadherin through the transcription factors SNAI1 and ERG, leading to dysfunction of glomerular filtration barrier and induction of albuminuria. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Liqun Li
- Institute of Microvascular Medicine, Medical Research Center (L.L., Q.L., J.L.), Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.,School of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China (L.L., T.S., W.S., X.W.)
| | - Qiang Liu
- Institute of Microvascular Medicine, Medical Research Center (L.L., Q.L., J.L.), Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Tongyao Shang
- School of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China (L.L., T.S., W.S., X.W.)
| | - Wei Song
- School of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China (L.L., T.S., W.S., X.W.)
| | - Dongmei Xu
- Department of Nephrology (D.X.), Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Thaddeus D Allen
- Molecular and Cellular Biology Division, Sunnybrook Health Science Centre (T.D.A., J.J., C.G.L.), University of Toronto, Ontario, Canada.,Department of Medical Biophysics (T.D.A., C.G.L.), University of Toronto, Ontario, Canada.,Tradewind BioScience, Daly City, California (T.D.A.)
| | - Xia Wang
- School of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China (L.L., T.S., W.S., X.W.)
| | - James Jeong
- General Internal Medicine, Markham Stouffville Hospital, Toronto, Ontario, Canada (J.J.)
| | - Corrinne G Lobe
- Molecular and Cellular Biology Division, Sunnybrook Health Science Centre (T.D.A., J.J., C.G.L.), University of Toronto, Ontario, Canada.,Department of Medical Biophysics (T.D.A., C.G.L.), University of Toronto, Ontario, Canada
| | - Ju Liu
- Institute of Microvascular Medicine, Medical Research Center (L.L., Q.L., J.L.), Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
12
|
Choi SR, Yang Y, Huang KY, Kong HJ, Flick MJ, Han B. Engineering of biomaterials for tumor modeling. MATERIALS TODAY. ADVANCES 2020; 8:100117. [PMID: 34541484 PMCID: PMC8448271 DOI: 10.1016/j.mtadv.2020.100117] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Development of biomaterials mimicking tumor and its microenvironment has recently emerged for the use of drug discovery, precision medicine, and cancer biology. These biomimetic models have developed by reconstituting tumor and stroma cells within the 3D extracellular matrix. The models are recently extended to recapitulate the in vivo tumor microenvironment, including biological, chemical, and mechanical conditions tailored for specific cancer type and its microenvironment. In spite of the recent emergence of various innovative engineered tumor models, many of these models are still early stage to be adapted for cancer research. In this article, we review the current status of biomaterials engineering for tumor models considering three main aspects - cellular engineering, matrix engineering, and engineering for microenvironmental conditions. Considering cancer-specific variability in these aspects, our discussion is focused on pancreatic cancer, specifically pancreatic ductal adenocarcinoma (PDAC). In addition, we further discussed the current challenges and future opportunities to create reliable and relevant tumor models.
Collapse
Affiliation(s)
- Sae Rome Choi
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Yi Yang
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, and Blood Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Kai-Yu Huang
- Department of Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hyun Joon Kong
- Department of Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Matthew J. Flick
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, and Blood Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
- Weldon School of Biomedical Engineering and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
13
|
Abstract
Two BβN-domains of fibrinogen are formed by the N-terminal portions of its two Bβ chains including amino acid residues Bβ1-65. Although their folding status is not well understood and the recombinant disulfide-linked (Bβ1-66)2 fragment corresponding to a pair of these domains was found to be unfolded, some data suggest that these domains may be folded in the parent molecule. In contrast, their major functional properties are well established. Removal of fibrinopeptides B (amino acid residues Bβ1-14) from these domains upon fibrinogen to fibrin conversion results in the exposure of multiple binding sites in fibrin βN-domains (residues β15-65). These sites provide interactions of the βN-domains with different proteins and cells and their participation in various physiological and pathological processes including fibrin assembly, fibrin-dependent angiogenesis, and fibrin-dependent leukocyte transmigration and thereby inflammation. The major goal of the present review is to summarize current view on the structure and function of these domains in fibrinogen and fibrin and their role in the above-mentioned processes.
Collapse
Affiliation(s)
- Leonid Medved
- Center for Vascular and Inflammatory Diseases and Departments of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, U.S.A
| | - Sergiy Yakovlev
- Center for Vascular and Inflammatory Diseases and Departments of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, U.S.A
| |
Collapse
|
14
|
Aggregation of platelets, proliferation of endothelial cells and motility of cancer cells are mediated by the B?1(15)-42 residue of fibrin(ogen). UKRAINIAN BIOCHEMICAL JOURNAL 2020. [DOI: 10.15407/ubj92.02.072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
15
|
Yakovlev S, Cao C, Galisteo R, Zhang L, Strickland DK, Medved L. Fibrin-VLDL Receptor-Dependent Pathway Promotes Leukocyte Transmigration by Inhibiting Src Kinase Fyn and is a Target for Fibrin β15-42 Peptide. Thromb Haemost 2019; 119:1816-1826. [PMID: 31466086 DOI: 10.1055/s-0039-1695008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
According to the current view, binding of fibrin degradation product E1 fragment to endothelial VE-cadherin promotes transendothelial migration of leukocytes and thereby inflammation, and fibrin-derived β15-42 peptide reduces leukocyte transmigration by competing with E1 for binding to VE-cadherin and, in addition, by signaling through Src kinase Fyn. However, the very low affinity of β15-42 to VE-cadherin raised a question about its ability to inhibit E1-VE-cadherin interaction. Further, our previous study revealed that fibrin promotes leukocyte transmigration through the very-low-density lipoprotein (VLDL) receptor (VLDLR)-dependent pathway and suggested a possible link between the inhibitory properties of β15-42 and this pathway. To test such a link and the proposed inhibitory mechanisms for β15-42, we performed in vitro experiments using surface plasmon resonance, enzyme-linked immunosorbent assay, and leukocyte transendothelial migration assay, and in vivo studies with wild-type and VLDLR-deficient mice using mouse model of peritonitis. The experiments revealed that β15-42 cannot inhibit E1-VE-cadherin interaction at the concentrations used in the previous in vivo studies leaving the proposed Fyn-dependent signaling mechanism as a viable explanation for the inhibitory effect of β15-42. While testing this mechanism, we confirmed that Fyn plays a critical role in controlling fibrin-induced transendothelial migration of leukocytes and found that signaling through the VLDLR-dependent pathway results in inhibition of Fyn, thereby increasing leukocyte transmigration. Furthermore, our in vivo experiments revealed that β15-42 inhibits this pathway, thereby preventing inhibition of Fyn and reducing leukocyte transmigration. Thus, this study clarifies the molecular mechanism underlying the VLDLR-dependent pathway of leukocyte transmigration and reveals that this pathway is a target for β15-42.
Collapse
Affiliation(s)
- Sergiy Yakovlev
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States.,Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Chunzhang Cao
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Rebeca Galisteo
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Li Zhang
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States.,Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Leonid Medved
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States.,Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
16
|
Golanov EV, Sharpe MA, Regnier-Golanov AS, Del Zoppo GJ, Baskin DS, Britz GW. Fibrinogen Chains Intrinsic to the Brain. Front Neurosci 2019; 13:541. [PMID: 31191233 PMCID: PMC6549596 DOI: 10.3389/fnins.2019.00541] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/09/2019] [Indexed: 11/13/2022] Open
Abstract
We observed fine fibrin deposition along the paravascular spaces in naive animals, which increased dramatically following subarachnoid hemorrhage (SAH). Following SAH, fibrin deposits in the areas remote from the hemorrhage. Traditionally it is thought that fibrinogen enters subarachnoid space through damaged blood brain barrier. However, deposition of fibrin remotely from hemorrhage suggests that fibrinogen chains Aα, Bβ, and γ can originate in the brain. Here we demonstrate in vivo and in vitro that astroglia and neurons are capable of expression of fibrinogen chains. SAH in mice was induced by the filament perforation of the circle of Willis. Four days after SAH animals were anesthetized, transcardially perfused and fixed. Whole brain was processed for immunofluorescent (IF) analysis of fibrin deposition on the brain surface or in brains slices processed for fibrinogen chains Aα, Bβ, γ immunohistochemical detection. Normal human astrocytes were grown media to confluency and stimulated with NOC-18 (100 μM), TNF-α (100 nM), ATP-γ-S (100 μM) for 24 h. Culture was fixed and washed/permeabilized with 0.1% Triton and processed for IF. Four days following SAH fibrinogen chains Aα IF associated with glia limitans and superficial brain layers increased 3.2 and 2.5 times (p < 0.05 and p < 0.01) on the ventral and dorsal brain surfaces respectively; fibrinogen chains Bβ increased by 3 times (p < 0.01) on the dorsal surface and fibrinogen chain γ increased by 3 times (p < 0.01) on the ventral surface compared to sham animals. Human cultured astrocytes and neurons constitutively expressed all three fibrinogen chains. Their expression changed differentially when exposed for 24 h to biologically significant stimuli: TNFα, NO or ATP. Western blot and RT-qPCR confirmed presence of the products of the appropriate molecular weight and respective mRNA. We demonstrate for the first time that mouse and human astrocytes and neurons express fibrinogen chains suggesting potential presence of endogenous to the brain fibrinogen chains differentially changing to biologically significant stimuli. SAH is followed by increased expression of fibrinogen chains associated with glia limitans remote from the hemorrhage. We conclude that brain astrocytes and neurons are capable of production of fibrinogen chains, which may be involved in various normal and pathological processes.
Collapse
Affiliation(s)
- Eugene V Golanov
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Martyn A Sharpe
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | | | - Gregory J Del Zoppo
- Division of Hematology, University of Washington School of Medicine, Seattle, WA, United States
| | - David S Baskin
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Gavin W Britz
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
17
|
Choi SM, Chaudhry P, Zo SM, Han SS. Advances in Protein-Based Materials: From Origin to Novel Biomaterials. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1078:161-210. [PMID: 30357624 DOI: 10.1007/978-981-13-0950-2_10] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Biomaterials play a very important role in biomedicine and tissue engineering where they directly affect the cellular activities and their microenvironment . Myriad of techniques have been employed to fabricate a vast number natural, artificial and recombinant polymer s in order to harness these biomaterials in tissue regene ration , drug delivery and various other applications. Despite of tremendous efforts made in this field during last few decades, advanced and new generation biomaterials are still lacking. Protein based biomaterials have emerged as an attractive alternatives due to their intrinsic properties like cell to cell interaction , structural support and cellular communications. Several protein based biomaterials like, collagen , keratin , elastin , silk protein and more recently recombinant protein s are being utilized in a number of biomedical and biotechnological processes. These protein-based biomaterials have enormous capabilities, which can completely revolutionize the biomaterial world. In this review, we address an up-to date review on the novel, protein-based biomaterials used for biomedical field including tissue engineering, medical science, regenerative medicine as well as drug delivery. Further, we have also emphasized the novel fabrication techniques associated with protein-based materials and implication of these biomaterials in the domain of biomedical engineering .
Collapse
Affiliation(s)
- Soon Mo Choi
- Regional Research Institute for Fiber&Fashion Materials, Yeungnam University, Gyeongsan, South Korea
| | - Prerna Chaudhry
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Sun Mi Zo
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea.
| |
Collapse
|
18
|
Yakovlev S, Medved L. Interaction of Fibrin with the Very Low-Density Lipoprotein (VLDL) Receptor: Further Characterization and Localization of the VLDL Receptor-Binding Site in Fibrin βN-Domains. Biochemistry 2017; 56:2518-2528. [PMID: 28437098 DOI: 10.1021/acs.biochem.7b00087] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Our recent study revealed that fibrin and the very low-density lipoprotein receptor (VLDLR) interact with each other through a pair of fibrin βN-domains and CR domains of the receptor and this interaction promotes transendothelial migration of leukocytes and thereby inflammation. The major objectives of this study were to further clarify the molecular mechanism of fibrin-VLDLR interaction and to identify amino acid residues in the βN-domains involved in this interaction. Our binding experiments with the (β15-66)2 fragment, which corresponds to a pair of fibrin βN-domains, and the VLDLR(1-8) fragment, consisting of eight CR domains of VLDLR, revealed that interaction between them strongly depends on ionic strength and chemical modification of all Lys or Arg residues in (β15-66)2 results in abrogation of this interaction. To identify which of these residues are involved in the interaction, we mutated all Lys or Arg residues in each of the three positively charged Lys/Arg clusters of the (β15-66)2 fragment, as well as single Arg17 and Arg30, and tested the affinity of the mutants obtained for VLDLR(1-8) by an enzyme-linked immunosorbent assay and surface plasmon resonance. The experiments revealed that the second and third Lys/Arg clusters make the major contribution to this interaction while the contribution of the first cluster is moderate. The results obtained suggest that interaction between fibrin and the VLDL receptor employs the "double-Lys/Arg" recognition mode previously proposed for the interaction of the LDL receptor family members with their ligands. They also provide valuable information for the development of highly specific peptide-based inhibitors of fibrin-VLDLR interaction.
Collapse
Affiliation(s)
- Sergiy Yakovlev
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine , Baltimore, Maryland 21201, United States
| | - Leonid Medved
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine , Baltimore, Maryland 21201, United States
| |
Collapse
|
19
|
Yakovlev S, Medved L. Interaction of Fibrin with the Very Low Density Lipoprotein Receptor: Further Characterization and Localization of the Fibrin-Binding Site. Biochemistry 2015; 54:4751-61. [PMID: 26153297 DOI: 10.1021/acs.biochem.5b00582] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Our recent study revealed that fibrin interacts with the very low density lipoprotein receptor (VLDLR) on endothelial cells through its βN domains, and this interaction promotes transendothelial migration of leukocytes and thereby inflammation. The major aims of this study were to further characterize this interaction and localize the fibrin-binding site in the VLDLR. To localize the fibrin-binding site, we expressed a soluble extracellular portion of this receptor, sVLDLRHT, its N- and C-terminal regions, VLDLR(1-8)HT and des(1-8)VLDLRHT, respectively, and a number of VLDLR fragments containing various combinations of CR domains and confirmed their proper folding by fluorescence spectroscopy. Interaction of these fragments with the (β15-66)2 fragment corresponding to a pair of VLDLR-binding βN domains of fibrin was tested by different methods. Our experiments performed by an enzyme-linked immunosorbent assay and surface plasmon resonance revealed that the VLDLR(1-8)HT fragment containing eight CR domains of VLDLR and its subfragments, VLDLR(1-4)HT and VLDLR(2-4)HT, interact with (β15-66)2 with practically the same affinity as sVLDLRHT while the affinity of VLDLR(2-3)HT was ∼2-fold lower. In contrast, des(1-8)VLDLRHT exhibited no binding. Formation of the complex in solution between the fibrin-binding fragments of VLDLR and (β15-66)2 was detected by fluorescence spectroscopy. In addition, formation of a complex between VLDLR(2-4)HT and (β15-66)2 in solution was confirmed by size-exclusion chromatography. Thus, the results obtained indicate that minimal fibrin-binding structures are located within the second and third CR domains of the VLDL receptor and the presence of the fourth CR domain is required for high-affinity binding. They also indicate that tryptophan residues of CR domains are involved in this binding.
Collapse
Affiliation(s)
- Sergiy Yakovlev
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Leonid Medved
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| |
Collapse
|
20
|
Angiogenic growth factors interactome and drug discovery: The contribution of surface plasmon resonance. Cytokine Growth Factor Rev 2014; 26:293-310. [PMID: 25465594 DOI: 10.1016/j.cytogfr.2014.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 11/10/2014] [Accepted: 11/11/2014] [Indexed: 11/21/2022]
Abstract
Angiogenesis is implicated in several pathological conditions, including cancer, and in regenerative processes, including the formation of collateral blood vessels after stroke. Physiological angiogenesis is the outcome of a fine balance between the action of angiogenic growth factors (AGFs) and anti-angiogenic molecules, while pathological angiogenesis occurs when this balance is pushed toward AGFs. AGFs interact with multiple endothelial cell (EC) surface receptors inducing cell proliferation, migration and proteases upregulation. On the contrary, free or extracellular matrix-associated molecules inhibit angiogenesis by sequestering AGFs (thus hampering EC stimulation) or by interacting with specific EC receptors inducing apoptosis or decreasing responsiveness to AGFs. Thus, angiogenesis results from an intricate network of interactions among pro- and anti-angiogenic molecules, EC receptors and various modulators. All these interactions represent targets for the development of pro- or anti-angiogenic therapies. These aims call for suitable technologies to study the countless interactions occurring during neovascularization. Surface plasmon resonance (SPR) is a label-free optical technique to study biomolecular interactions in real time. It has become the golden standard technology for interaction analysis in biomedical research, including angiogenesis. From a survey of the literature it emerges that SPR has already contributed substantially to the better understanding of the neovascularization process, laying the basis for the decoding of the angiogenesis "interactome" and the identification of "hub molecules" that may represent preferential targets for an efficacious modulation of angiogenesis. Here, the still unexploited full potential of SPR is enlightened, pointing to improvements in its use for a deeper understanding of the mechanisms of neovascularization and the identification of novel anti-angiogenic drugs.
Collapse
|
21
|
Ceccarelli J, Putnam AJ. Sculpting the blank slate: how fibrin's support of vascularization can inspire biomaterial design. Acta Biomater 2014; 10:1515-23. [PMID: 23933102 DOI: 10.1016/j.actbio.2013.07.043] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 07/19/2013] [Accepted: 07/31/2013] [Indexed: 12/12/2022]
Abstract
Fibrin is the primary extracellular constituent of blood clots, and plays an important role as a provisional matrix during wound healing and tissue remodeling. Fibrin-based biomaterials have proven their utility as hemostatic therapies, scaffolds for tissue engineering, vehicles for controlled release, and platforms for culturing and studying cells in three dimensions. Nevertheless, fibrin presents a complex milieu of signals to embedded cells, many of which are not well understood. Synthetic extracellular matrices (ECMs) provide a blank slate that can ostensibly be populated with specific bioactive cues, including growth factors, growth factor binding motifs, adhesive peptides and peptide crosslinks susceptible to proteases, thereby enabling a degree of customization for specific applications. However, the continued evolution and improvement of synthetic ECMs requires parallel efforts to deconstruct native ECMs and decipher the cues they provide to constituent cells. The objective of this review is to reintroduce fibrin, a protein with a well-characterized structure and biochemistry, and its ability to support angiogenesis specifically. Although fibrin's structure-function relationships have been studied for decades, opportunities to engineer new and improved synthetic hydrogels can be realized by further exploiting fibrin's inspiring design.
Collapse
Affiliation(s)
- Jacob Ceccarelli
- Department of Biomedical Engineering, University of Michigan, 2154 Lurie Biomedical Engineering Building, 1101 Beal Ave, Ann Arbor, MI 48109, USA
| | - Andrew J Putnam
- Department of Biomedical Engineering, University of Michigan, 2154 Lurie Biomedical Engineering Building, 1101 Beal Ave, Ann Arbor, MI 48109, USA.
| |
Collapse
|
22
|
Messina PV, Hassan N, Soltero A, Ruso JM. Identifying emerging trends of protein hydrogels for biological scaffolding. RSC Adv 2013. [DOI: 10.1039/c3ra42204f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
23
|
Wang C, Li Y. Interaction and nanotoxic effect of TiO₂ nanoparticle on fibrinogen by multi-spectroscopic method. THE SCIENCE OF THE TOTAL ENVIRONMENT 2012; 429:156-160. [PMID: 22607744 DOI: 10.1016/j.scitotenv.2012.03.048] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 03/20/2012] [Accepted: 03/20/2012] [Indexed: 06/01/2023]
Abstract
Toxicological effects of nanoparticles (NPs) are still poorly documented while there are great demands for industrial applications and daily life. The aim of this study is to evaluate the influence of physicochemical characteristics on TiO₂ NP toxicological effects toward protein. In order to better understand the physicochemical basis of the toxic of NP in industrial applications and under conditions of environmental exposure, we performed an array of photophysical measurements to quantify the interaction of TiO₂ NP with protein. Fluorescence quenching, circular dichroism, dynamic light scattering and transmission electron microscopy measurements were performed on TiO₂ NP having a diameter range from 10 to 35 nm in the performance of protein. We find that the TiO₂ NP strongly associates with protein where the binding constant, as well as the degree of cooperativity of particle-protein binding, depends on particle size. We also find tentative evidence that the protein undergoes conformational change upon association with the NP. These results indicate that exposure to TiO₂ NP may have an unfavorable effect on human health by inactivating functional proteins.
Collapse
Affiliation(s)
- Chao Wang
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lakes of Ministry of Education, College of Environment, HoHai University, NanJing, JiangSu Province 210098, China
| | | |
Collapse
|
24
|
Rich RL, Myszka DG. Survey of the 2009 commercial optical biosensor literature. J Mol Recognit 2012; 24:892-914. [PMID: 22038797 DOI: 10.1002/jmr.1138] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We took a different approach to reviewing the commercial biosensor literature this year by inviting 22 biosensor users to serve as a review committee. They set the criteria for what to expect in a publication and ultimately decided to use a pass/fail system for selecting which papers to include in this year's reference list. Of the 1514 publications in 2009 that reported using commercially available optical biosensor technology, only 20% passed their cutoff. The most common criticism the reviewers had with the literature was that "the biosensor experiments could have been done better." They selected 10 papers to highlight good experimental technique, data presentation, and unique applications of the technology. This communal review process was educational for everyone involved and one we will not soon forget.
Collapse
Affiliation(s)
- Rebecca L Rich
- Center for Biomolecular Interaction Analysis, University of Utah, Salt Lake City, UT, USA
| | | |
Collapse
|
25
|
Identification of VLDLR as a novel endothelial cell receptor for fibrin that modulates fibrin-dependent transendothelial migration of leukocytes. Blood 2011; 119:637-44. [PMID: 22096238 DOI: 10.1182/blood-2011-09-382580] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
While testing the effect of the (β15-66)(2) fragment, which mimics a pair of fibrin βN-domains, on the morphology of endothelial cells, we found that this fragment induces redistribution of vascular endothelial-cadherin in a process that is inhibited by the receptor-associated protein (RAP). Based on this finding, we hypothesized that fibrin may interact with members of RAP-dependent low-density lipoprotein (LDL) receptor family. To test this hypothesis, we examined the interaction of (β15-66)(2), fibrin, and several fibrin-derived fragments with 2 members of this family by ELISA and surface plasmon resonance. The experiments showed that very LDL (VLDL) receptor (VLDLR) interacts with high affinity with fibrin through its βN-domains, and this interaction is inhibited by RAP and (β15-66)(2). Furthermore, RAP inhibited transendothelial migration of neutrophils induced by fibrin-derived NDSK-II fragment containing βN-domains, suggesting the involvement of VLDLR in fibrin-dependent leukocyte transmigration. Our experiments with VLDLR-deficient mice confirmed this suggestion by showing that, in contrast to wild-type mice, fibrin-dependent leukocyte transmigration does not occur in such mice. Altogether, the present study identified VLDLR as a novel endothelial cell receptor for fibrin that promotes fibrin-dependent leukocyte transmigration and thereby inflammation. Establishing the molecular mechanism underlying this interaction may result in the development of novel inhibitors of fibrin-dependent inflammation.
Collapse
|
26
|
|
27
|
YAKOVLEV S, GAO Y, CAO C, CHEN L, STRICKLAND DK, ZHANG L, MEDVED L. Interaction of fibrin with VE-cadherin and anti-inflammatory effect of fibrin-derived fragments. J Thromb Haemost 2011; 9:1847-55. [PMID: 21752185 PMCID: PMC3166367 DOI: 10.1111/j.1538-7836.2011.04438.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND The interaction of the fibrin βN-domain with VE-cadherin on endothelial cells is implicated in transendothelial migration of leukocytes, and the β15-42 fragment representing part of this domain has been shown to inhibit this process. However, our previous study revealed that only a dimeric (β15-66)(2) fragment, corresponding to the full-length βN-domain and mimicking its dimeric arrangement in fibrin, bound to VE-cadherin. OBJECTIVE To test our hypothesis that dimerization of β15-42-containing fragments increases their affinity for VE-cadherin and ability to inhibit transendothelial migration of leukocytes. METHODS Interaction of β15-42-containing fragments with VE-cadherin was characterized by ELISA and surface plasmon resonance. The inhibitory effect of such fragments was tested in vitro with a leukocyte transendothelial migration assay and in vivo with mouse models of peritonitis and myocardial ischemia-reperfusion injury. RESULTS First, we prepared the monomeric β15-42 and β15-64 fragments and their dimeric forms, (β15-44)(2) and (β15-66)(2) , and studied their interaction with the fibrin-binding domain of VE-cadherin, VE-cad(3). The experiments revealed that both dimeric fragments bound to VE-cad(3) with high affinity, whereas the affinities of β15-42 and β15-64 were significantly lower. Next, we tested the ability of these fragments to inhibit leukocyte transmigration in vitro and infiltration into the inflamed peritoneum in vivo, and found that the inhibitory effects of the dimers on these processes were also superior. Furthermore, (β15-44)(2) significantly reduced myocardial injury induced by ischemia-reperfusion. CONCLUSION The results confirm our hypotheses and indicate that (β15-66)(2) and (β15-44)(2) , which exhibited much higher affinity for VE-cadherin, are highly effective in suppressing inflammation by inhibiting leukocyte transmigration.
Collapse
Affiliation(s)
- S. YAKOVLEV
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Y. GAO
- Center for Vascular and Inflammatory Diseases and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - C. CAO
- Center for Vascular and Inflammatory Diseases and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - L. CHEN
- Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - D. K. STRICKLAND
- Center for Vascular and Inflammatory Diseases and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - L. ZHANG
- Center for Vascular and Inflammatory Diseases and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - L. MEDVED
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
28
|
Hassan N, Maldonado-Valderrama J, Gunning AP, Morris VJ, Ruso JM. Investigating the effect of an arterial hypertension drug on the structural properties of plasma protein. Colloids Surf B Biointerfaces 2011; 87:489-97. [PMID: 21726985 DOI: 10.1016/j.colsurfb.2011.06.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/02/2011] [Accepted: 06/09/2011] [Indexed: 11/26/2022]
Abstract
Propanolol is a betablocker drug used in the treatment of arterial hypertension related diseases. In order to achieve an optimal performance of this drug it is important to consider the possible interactions of propanolol with plasma proteins. In this work, we have used several experimental techniques to characterise the effect of addition of the betablocker propanolol on the properties of bovine plasma fibrinogen (FB). Differential scanning calorimeter (DSC), circular dichroism (CD), dynamic light scattering (DLS), surface tension techniques and atomic force microscopy (AFM) measurements have been combined to carry out a detailed physicochemical and surface characterization of the mixed system. As a result, DSC measurements show that propranolol can play two opposite roles, either acting as a structure stabilizer at low molar concentrations or as a structure destabilizer at higher concentrations, in different domains of fibrinogen. CD measurements have revealed that the effect of propanolol on the secondary structure of fibrinogen depends on the temperature and the drug concentration and the DLS analysis showed evidence for protein aggregation. Interestingly, surface tension measurements provided further evidence of the conformational change induced by propanolol on the secondary structure of FB by importantly increasing the surface tension of the system. Finally, AFM imaging of the fibrinogen system provided direct visualization of the protein structure in the presence of propanolol. Combination of these techniques has produced complementary information on the behavior of the mixed system, providing new insights into the structural properties of proteins with potential medical interest.
Collapse
Affiliation(s)
- Natalia Hassan
- Department of Applied Physics University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | | | | |
Collapse
|
29
|
Hassan N, Barbosa LRS, Itri R, Ruso JM. Fibrinogen stability under surfactant interaction. J Colloid Interface Sci 2011; 362:118-26. [PMID: 21722913 DOI: 10.1016/j.jcis.2011.06.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 06/02/2011] [Accepted: 06/02/2011] [Indexed: 10/18/2022]
Abstract
Differential scanning calorimetry (DSC), circular dichroism (CD), difference spectroscopy (UV-vis), Raman spectroscopy, and small-angle X-ray scattering (SAXS) measurements have been performed in the present work to provide a quantitatively comprehensive physicochemical description of the complexation between bovine fibrinogen and the sodium perfluorooctanoate, sodium octanoate, and sodium dodecanoate in glycine buffer (pH 8.5). It has been found that sodium octanoate and dodecanoate act as fibrinogen destabilizer. Meanwhile, sodium perfluorooctanoate acts as a structure stabilizer at low molar concentration and as a destabilizer at high molar concentration. Fibrinogen's secondary structure is affected by all three studied surfactants (decrease in α-helix and an increase in β-sheet content) to a different extent. DSC and UV-vis revealed the existence of intermediate states in the thermal unfolding process of fibrinogen. In addition, SAXS data analysis showed that pure fibrinogen adopts a paired-dimer structure in solution. Such a structure is unaltered by sodium octanoate and perfluoroctanoate. However, interaction of sodium dodecanoate with the fibrinogen affects the protein conformation leading to a complex formation. Taken together, all results evidence that both surfactant hydrophobicity and tail length mediate the fibrinogen stability upon interaction.
Collapse
Affiliation(s)
- Natalia Hassan
- Soft Matter and Molecular Biophysics Group, Department of Applied Physics, University of Santiago de Compostela, Campus Vida s/n, 15782 Santiago de Compostela, Spain
| | | | | | | |
Collapse
|
30
|
Hassan N, Ruso JM, Somasundaran P. Mechanisms of fibrinogen–acebutolol interactions: Insights from DSC, CD and LS. Colloids Surf B Biointerfaces 2011; 82:581-7. [DOI: 10.1016/j.colsurfb.2010.10.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 09/06/2010] [Accepted: 10/11/2010] [Indexed: 11/25/2022]
|
31
|
Abstract
PURPOSE OF REVIEW Proper control of endothelial cell contacts is the basis for maintenance of the vascular barrier function. Loss of this function leads to leak of fluid and protein from the vasculature and extensive leaks cause shock and death. The endothelial barrier also controls the entry of leukocytes into tissue and it is believed that leukocytes target endothelial cell contacts to reach sites of inflammation. RECENT FINDINGS Within the last 2 years several new molecular players and molecular interactions have been identified that either help in stabilizing the endothelial contacts or mediate their opening if triggered by the appropriate stimuli. Novel signaling mechanisms have been identified that regulate endothelial cell contacts. Whether, how and to what extent the complex of the endothelial specific adhesion molecule vascular endothelial-cadherin and its associated catenins is involved in these processes will be a major focus of this article. SUMMARY Endothelial cell contacts are regulated by a complex interplay between various receptors and signaling mediators that control the plasticity of the cytoskeleton and the function of junctional adhesion molecules. Knowing and understanding the essential players of this network will allow development of agents that could prevent breakdown of the vascular permeability barrier in shock or that could block leukocyte extravasation and thereby antagonize inflammation.
Collapse
|