1
|
Wang Q, Zhao X, Yu F, Fang PH, Liu L, Du X, Li W, He D, Bai Y, Li S, Yuan J. Photocurable and Temperature-Sensitive Bioadhesive Hydrogels for Sutureless Sealing of Full-Thickness Corneal Wounds. SMALL METHODS 2024; 8:e2300996. [PMID: 37997553 DOI: 10.1002/smtd.202300996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/19/2023] [Indexed: 11/25/2023]
Abstract
Penetrating corneal wounds can cause severe vision impairment and require prompt intervention to restore globe integrity and minimize the risk of infection. Tissue adhesives have emerged as a promising alternative to suturing for mitigating postoperative complications. However, conventional water-soluble adhesives suffer formidable challenges in sealing penetrating corneal wounds due to dilution or loss in a moist environment. Inspired by the robust adhesion of mussels in aquatic conditions, an injectable photocurable bioadhesive hydrogel (referred to as F20HD5) composed of polyether F127 diacrylate and dopamine-modified hyaluronic acid methacrylate is developed for sutureless closure of corneal full-thickness wounds. F20HD5 exhibits high transparency, wound-sealing ability, proper viscosity, biodegradability, and excellent biocompatibility. It allows in situ cross-linking via visible light, thereby providing sufficient mechanical strength and adhesiveness. In vivo, the adhesive hydrogel effectively closed penetrating linear corneal incisions and corneal injuries with minimal tissue loss in rabbits. During the 56-day follow-up, the hydrogel facilitates the repair of the injured corneas, resulting in more symmetrical curvatures and less scarring in distinction to the untreated control. Thus, bioinspired hydrogel holds promise as an effective adhesive for sealing full-thickness corneal wounds.
Collapse
Affiliation(s)
- Qian Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Xuan Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Fei Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Po-Han Fang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Liu Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Xinyue Du
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Weihua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Dalian He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Ying Bai
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, SunYat-sen University, Guangzhou, 510006, China
| | - Saiqun Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Jin Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| |
Collapse
|
2
|
Message in a Scaffold: Natural Biomaterials for Three-Dimensional (3D) Bioprinting of Human Brain Organoids. Biomolecules 2022; 13:biom13010025. [PMID: 36671410 PMCID: PMC9855696 DOI: 10.3390/biom13010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/07/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Brain organoids are invaluable tools for pathophysiological studies or drug screening, but there are still challenges to overcome in making them more reproducible and relevant. Recent advances in three-dimensional (3D) bioprinting of human neural organoids is an emerging approach that may overcome the limitations of self-organized organoids. It requires the development of optimal hydrogels, and a wealth of research has improved our knowledge about biomaterials both in terms of their intrinsic properties and their relevance on 3D culture of brain cells and tissue. Although biomaterials are rarely biologically neutral, few articles have reviewed their roles on neural cells. We here review the current knowledge on unmodified biomaterials amenable to support 3D bioprinting of neural organoids with a particular interest in their impact on cell homeostasis. Alginate is a particularly suitable bioink base for cell encapsulation. Gelatine is a valuable helper agent for 3D bioprinting due to its viscosity. Collagen, fibrin, hyaluronic acid and laminin provide biological support to adhesion, motility, differentiation or synaptogenesis and optimize the 3D culture of neural cells. Optimization of specialized hydrogels to direct differentiation of stem cells together with an increased resolution in phenotype analysis will further extend the spectrum of possible bioprinted brain disease models.
Collapse
|
3
|
Growth of MIN-6 Cells on Salmon Fibrinogen Scaffold Improves Insulin Secretion. Pharmaceutics 2022; 14:pharmaceutics14050941. [PMID: 35631527 PMCID: PMC9144899 DOI: 10.3390/pharmaceutics14050941] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 02/04/2023] Open
Abstract
The incidence of type I diabetes has been increasing worldwide at an annual rate of approximately 3%. One of the strategies to treat type I diabetes is islet transplantation, in which damaged β-cells are replaced with new islets. To improve β-cells’ expansion and pseudoislet formation, studies are focusing on using extracellular-matrix-resembling substrates. We evaluated the potential of salmon fibrinogen and chitosan electrospun scaffold as cell substrate for cultivating MIN-6 cells. The morphology of cells, insulin secretion and gene expression was evaluated and compared with other substrates (nanofibrous scaffold, microporous scaffold and tissue culture polystyrene). We found that all tested 3D conditions favored the pseudoislet formation of MIN-6 cells. The insulin secretion of MIN-6 cells after stimulation with high-glucose media shows approximately a 9-fold increase compared to the control group when a fibrinogen/chitosan-based electrospun scaffold was used for cultivation. The differences in insulin secretion were corroborated by differences in gene expression. The differences in insulin secretion could probably be attributed to the differences in the mechanical and/or chemical nature of the tested substrates.
Collapse
|
4
|
Barrows CM, Wu D, Farach-Carson MC, Young S. Building a Functional Salivary Gland for Cell-Based Therapy: More than Secretory Epithelial Acini. Tissue Eng Part A 2020; 26:1332-1348. [PMID: 32829674 PMCID: PMC7759264 DOI: 10.1089/ten.tea.2020.0184] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/20/2020] [Indexed: 11/13/2022] Open
Abstract
A few treatment options exist for patients experiencing xerostomia due to hyposalivation that occurs as a result of disease or injury to the gland. An opportunity for a permanent solution lies in the field of salivary gland replacement through tissue engineering. Recent success emboldens in the vision of producing a tissue-engineered salivary gland composed of differentiated salivary epithelial cells that are able to differentiate to form functional units that produce and deliver saliva to the oral cavity. This vision is augmented by advances in understanding cellular mechanisms that guide branching morphogenesis and salivary epithelial cell polarization in both acinar and ductal structures. Growth factors and other guidance cues introduced into engineered constructs help to develop a more complex glandular structure that seeks to mimic native salivary gland tissue. This review describes the separate epithelial phenotypes that make up the gland, and it describes their relationship with the other cell types such as nerve and vasculature that surround them. The review is organized around the links between the native components that form and contribute to various aspects of salivary gland development, structure, and function and how this information can drive the design of functional tissue-engineered constructs. In addition, we discuss the attributes of various biomaterials commonly used to drive function and form in engineered constructs. The review also contains a current description of the state-of-the-art of the field, including successes and challenges in creating materials for preclinical testing in animal models. The ability to integrate biomolecular cues in combination with a range of materials opens the door to the design of increasingly complex salivary gland structures that, once accomplished, can lead to breakthroughs in other fields of tissue engineering of epithelial-based exocrine glands or oral tissues.
Collapse
Affiliation(s)
- Caitlynn M.L. Barrows
- Department of Diagnostic and Biomedical Sciences and The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
- Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences and The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
| | - Mary C. Farach-Carson
- Department of Diagnostic and Biomedical Sciences and The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
- Department of Biosciences and Rice University, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Simon Young
- Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
| |
Collapse
|
5
|
Ibarra A, Mendieta-Arbesú E, Suarez-Meade P, García-Vences E, Martiñón S, Rodriguez-Barrera R, Lomelí J, Flores-Romero A, Silva-García R, Buzoianu-Anguiano V, Borlongan CV, Frydman TD. Motor Recovery after Chronic Spinal Cord Transection in Rats: A Proof-of-Concept Study Evaluating a Combined Strategy. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 18:52-62. [DOI: 10.2174/1871527317666181105101756] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/20/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022]
Abstract
Background:
The chronic phase of Spinal Cord (SC) injury is characterized by the presence
of a hostile microenvironment that causes low activity and a progressive decline in neurological function;
this phase is non-compatible with regeneration. Several treatment strategies have been investigated
in chronic SC injury with no satisfactory results. OBJECTIVE- In this proof-of-concept study,
we designed a combination therapy (Comb Tx) consisting of surgical glial scar removal plus scar inhibition,
accompanied with implantation of mesenchymal stem cells (MSC), and immunization with
neural-derived peptides (INDP).
Methods:
This study was divided into three subsets, all in which Sprague Dawley rats were subjected
to a complete SC transection. Sixty days after injury, animals were randomly allocated into two groups
for therapeutic intervention: control group and animals receiving the Comb-Tx. Sixty-three days after
treatment we carried out experiments analyzing motor recovery, presence of somatosensory evoked
potentials, neural regeneration-related genes, and histological evaluation of serotoninergic fibers.
Results:
Comb-Tx induced a significant locomotor and electrophysiological recovery. An increase in the
expression of regeneration-associated genes and the percentage of 5-HT+ fibers was noted at the caudal
stump of the SC of animals receiving the Comb-Tx. There was a significant correlation of locomotor recovery
with positive electrophysiological activity, expression of GAP43, and percentage of 5-HT+ fibers.
Conclusion:
Comb-Tx promotes motor and electrophysiological recovery in the chronic phase of SC
injury subsequent to a complete transection. Likewise, it is capable of inducing the permissive microenvironment
to promote axonal regeneration.
Collapse
Affiliation(s)
- Antonio Ibarra
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | - Erika Mendieta-Arbesú
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | - Paola Suarez-Meade
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | - Elisa García-Vences
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | | | - Roxana Rodriguez-Barrera
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | - Joel Lomelí
- Instituto Politecnico Nacional, Escuela Superior de Medicina, Ciudad de Mexico, Mexico
| | - Adrian Flores-Romero
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | | | | | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, United States
| | - Tamara D. Frydman
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| |
Collapse
|
6
|
Gačanin J, Hedrich J, Sieste S, Glaßer G, Lieberwirth I, Schilling C, Fischer S, Barth H, Knöll B, Synatschke CV, Weil T. Autonomous Ultrafast Self-Healing Hydrogels by pH-Responsive Functional Nanofiber Gelators as Cell Matrices. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805044. [PMID: 30411838 DOI: 10.1002/adma.201805044] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/19/2018] [Indexed: 06/08/2023]
Abstract
The synthesis of hybrid hydrogels by pH-controlled structural transition with exceptional rheological properties as cellular matrix is reported. "Depsi" peptide sequences are grafted onto a polypeptide backbone that undergo a pH-induced intramolecular O-N-acyl migration at physiological conditions affording peptide nanofibers (PNFs) as supramolecular gelators. The polypeptide-PNF hydrogels are mechanically remarkably robust. They reveal exciting thixotropic behavior with immediate in situ recovery after exposure to various high strains over long periods and self-repair of defects by instantaneous reassembly. High cytocompatibility, convenient functionalization by coassembly, and controlled enzymatic degradation but stability in 2D and 3D cell culture as demonstrated by the encapsulation of primary human umbilical vein endothelial cells and neuronal cells open many attractive opportunities for 3D tissue engineering and other biomedical applications.
Collapse
Affiliation(s)
- Jasmina Gačanin
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
- Institute of Inorganic Chemistry I, University of Ulm, 89081, Ulm, Germany
| | - Jana Hedrich
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Stefanie Sieste
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
- Institute of Inorganic Chemistry I, University of Ulm, 89081, Ulm, Germany
| | - Gunnar Glaßer
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Ingo Lieberwirth
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Corinna Schilling
- Institute of Physiological Chemistry, University of Ulm, 89081, Ulm, Germany
| | - Stephan Fischer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, 89081, Ulm, Germany
| | - Holger Barth
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, 89081, Ulm, Germany
| | - Bernd Knöll
- Institute of Physiological Chemistry, University of Ulm, 89081, Ulm, Germany
| | | | - Tanja Weil
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
- Institute of Inorganic Chemistry I, University of Ulm, 89081, Ulm, Germany
| |
Collapse
|
7
|
Laidmäe I, Ērglis K, Cēbers A, Janmey PA, Uibo R. Salmon fibrinogen and chitosan scaffold for tissue engineering: in vitro and in vivo evaluation. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2018; 29:182. [PMID: 30506370 PMCID: PMC6267118 DOI: 10.1007/s10856-018-6192-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 11/16/2018] [Indexed: 06/09/2023]
Abstract
3D fibrous scaffolds have received much recent attention in regenerative medicine. Use of fibrous scaffolds has shown promising results in tissue engineering and wound healing. Here we report the development and properties of a novel fibrous scaffold that is useful for promoting wound healing. A scaffold made of salmon fibrinogen and chitosan is produced by electrospinning, resulting in a biocompatible material mimicking the structure of the native extracellular matrix (ECM) with suitable biochemical and mechanical properties. The scaffold is produced without the need for enzymes, in particular thrombin, but is fully compatible with their addition if needed. Human dermal fibroblasts cultured on this scaffold showed progressive proliferation for 14 days. Split-thickness experimental skin wounds treated and untreated were compared in a 10-day follow-up period. Wound healing was more effective using the fibrinogen-chitosan scaffold than in untreated wounds. This scaffold could be applicable in various medical purposes including surgery, tissue regeneration, burns, traumatic injuries, and 3D cell culture platforms.
Collapse
Affiliation(s)
- Ivo Laidmäe
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411, Tartu, Estonia.
- Institute of Pharmacy, University of Tartu, 50411, Tartu, Estonia.
| | - Kaspars Ērglis
- Faculty of Physics, Mathematics and Optometry, University of Latvia, Riga, LV-1002, Latvia
| | - Andrejs Cēbers
- Faculty of Physics, Mathematics and Optometry, University of Latvia, Riga, LV-1002, Latvia
| | - Paul A Janmey
- Institute for Medicine and Engineering and Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Raivo Uibo
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411, Tartu, Estonia
| |
Collapse
|
8
|
Douglas SA, Lamothe SE, Singleton TS, Averett RD, Platt MO. Human cathepsins K, L, and S: Related proteases, but unique fibrinolytic activity. Biochim Biophys Acta Gen Subj 2018; 1862:1925-1932. [PMID: 29944896 DOI: 10.1016/j.bbagen.2018.06.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 06/06/2018] [Accepted: 06/19/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND Fibrin formation and dissolution are attributed to cascades of protease activation concluding with thrombin activation, and plasmin proteolysis for fibrin breakdown. Cysteine cathepsins are powerful proteases secreted by endothelial cells and others during cardiovascular disease and diabetes. Their fibrinolytic activity and putative role in hemostasis has not been well described. METHODS Fibrin gels were polymerized and incubated with recombinant human cathepsins (cat) K, L, or S, or plasmin, for dose-dependent and time-dependent studies. Dissolution of fibrin gels was imaged. SDS-PAGE was used to resolve cleaved fragments released from fibrin gels and remnant insoluble fibrin gel that was solubilized prior to electrophoresis to assess fibrin α, β, and γ polypeptide hydrolysis by cathepsins. Multiplex cathepsin zymography determined active amounts of cathepsins remaining. RESULTS There was significant loss of α and β fibrin polypeptides after incubation with cathepsins, with catS completely dissolving fibrin gel by 24 h. Binding to fibrin stabilized catL active time; it associated with cleaved fibrin fragments of multiple sizes. This was not observed for catK or S. CatS also remained active for longer times during fibrin incubation, but its association/binding did not withstand SDS-PAGE preparation. CONCLUSIONS Human cathepsins K, L, and S are fibrinolytic, and specifically can degrade the α and β fibrin polypeptide chains, generating fragments unique from plasmin. GENERAL SIGNIFICANCE Demonstration of cathepsins K, L, and S fibrinolytic activity leads to further investigation of contributory roles in disrupting vascular hemostasis, or breakdown of fibrin-based engineered vascular constructs where non-plasmin mediated fibrinolysis must be considered.
Collapse
Affiliation(s)
- Simone A Douglas
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology & Emory University, USA.
| | - Sarah E Lamothe
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology & Emory University, USA.
| | - Tatiyanna S Singleton
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology & Emory University, USA.
| | - Rodney D Averett
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, USA.
| | - Manu O Platt
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology & Emory University, USA.
| |
Collapse
|
9
|
Fibrin-Enhanced Canonical Wnt Signaling Directs Plasminogen Expression in Cementoblasts. Int J Mol Sci 2017; 18:ijms18112380. [PMID: 29120400 PMCID: PMC5713349 DOI: 10.3390/ijms18112380] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 11/17/2022] Open
Abstract
Cementum is a mineralized layer on the tooth's root surface and facilitates the biomechanical anchoring of fibrous connective tissues as a part of tooth-supportive complexes. Previously, we observed that OCCM30 cementoblasts cultured on fibrin matrices underwent apoptosis due to fibrin degradation through the expression of proteases. Here, we demonstrated that OCCM30 on fibrin matrices (OCCM30-fibrin) enhanced canonical Wnt signaling, which directed to plasminogen expression. The OCCM30-fibrin showed higher levels of Wnt3a expression, nuclear translocation of β-catenin, and T-cell factor (TCF) optimal motif (TOP) reporter activity than the cells on tissue culture dishes (OCCM30-TCD), indicating that the OCCM30-fibrin enhanced canonical Wnt/β-catenin signaling. Also, OCCM30-fibrin expressed biomineralization-associated markers at higher levels than OCCM30-TCD, of which levels were further increased with LiCl, a Wnt signaling activator. The OCCM30 cementoblasts simultaneously showed that high levels of plasminogen, a critical component of fibrinolysis, were expressed in the OCCM30-fibrin. Activation of canonical Wnt signaling with LiCl treatment or with forced lymphoid enhancer factor 1 (LEF1)-expression increased the expression of plasminogen. On the contrary, the inhibition of canonical Wnt signaling with siRNAs against Wnt3a or β-catenin abrogated fibrin-enhanced plasminogen expression. Furthermore, there are three conserved putative response elements for the LEF1/β-catenin complex in the plasminogen proximal promoter regions (-900 to +54). Site-directed mutations and chromatin immunoprecipitation indicated that canonical Wnt signaling directed plasminogen expression. Taken together, this study suggests that fibrin-based materials can modulate functional periodontal formations in controlling cementoblast differentiation and fibrin degradation.
Collapse
|
10
|
Dawn A, Kumari H. Low Molecular Weight Supramolecular Gels Under Shear: Rheology as the Tool for Elucidating Structure-Function Correlation. Chemistry 2017; 24:762-776. [PMID: 28952169 DOI: 10.1002/chem.201703374] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Indexed: 12/23/2022]
Abstract
Self-healing low molecular weight supramolecular gels (SMGs) represent an emerging class of smart materials, which can closely mimic the complex biological healing process, such as blood clotting, bone repair or wound healing. However, a lack of understanding of the structure-function correlation in the self-assembly process limits their molecular design and subsequent property tuning. The indispensability of a rheological study on supramolecular gels lies in direct transcription of the assembly property to the viscoelastic behavior of the material. This is similarly relevant to healable and non-healable systems. Thus, using rheology as a tool for elucidating structure-function relationships in self-assembled systems has huge potential. This review article will depict a general introduction of rheology in the field of soft matter including SMGs, followed by representative studies with interpretations, and discussion on future challenges. Altogether, this would be an effort, where an in-depth rheological study complemented with a real-time visualization with the help of microscopy, and introduction of other sophisticated real-time experiments, could be a step forward to capture the mystery of self-assembly process.
Collapse
Affiliation(s)
- Arnab Dawn
- James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
| | - Harshita Kumari
- James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
| |
Collapse
|
11
|
Park CH, Oh JH, Jung HM, Choi Y, Rahman SU, Kim S, Kim TI, Shin HI, Lee YS, Yu FH, Baek JH, Ryoo HM, Woo KM. Effects of the incorporation of ε-aminocaproic acid/chitosan particles to fibrin on cementoblast differentiation and cementum regeneration. Acta Biomater 2017; 61:134-143. [PMID: 28764948 DOI: 10.1016/j.actbio.2017.07.039] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/18/2017] [Accepted: 07/27/2017] [Indexed: 01/08/2023]
Abstract
Cementum formation on the exposed tooth-root surface is a critical process in periodontal regeneration. Although various therapeutic approaches have been developed, regeneration of integrated and functional periodontal complexes is still wanting. Here, we found that the OCCM30 cementoblasts cultured on fibrin matrix express substantial levels of matrix proteinases, leading to the degradation of fibrin and the apoptosis of OCCM30 cells, which was reversed upon treatment with a proteinase inhibitor, ε-aminocaproic acid (ACA). Based on these findings, ACA-releasing chitosan particles (ACP) were fabricated and ACP-incorporated fibrin (fibrin-ACP) promoted the differentiation of cementoblasts in vitro, as confirmed by bio-mineralization and expressions of molecules associated with mineralization. In a periodontal defect model of beagle dogs, fibrin-ACP resulted in substantial cementum formation on the exposed root dentin in vivo, compared to fibrin-only and enamel matrix derivative (EMD) which is used clinically for periodontal regeneration. Remarkably, the fibrin-ACP developed structural integrations of the cementum-periodontal ligament-bone complex by the Sharpey's fiber insertion. In addition, fibrin-ACP promoted alveolar bone regeneration through increased bone volume of tooth roof-of-furcation defects and root coverage. Therefore, fibrin-ACP can promote cementogenesis and osteogenesis by controlling biodegradability of fibrin, implicating the feasibility of its therapeutic use to improve periodontal regeneration. STATEMENT OF SIGNIFICANCE Cementum, the mineralized layer on root dentin surfaces, functions to anchor fibrous connective tissues on tooth-root surfaces with the collagenous Sharpey's fibers integration, of which are essential for periodontal functioning restoration in the complex. Through the cementum-responsible fiber insertions on tooth-root surfaces, PDLs transmit various mechanical responses to periodontal complexes against masticatory/occlusal stimulations to support teeth. In this study, periodontal tissue regeneration was enhanced by use of modified fibrin biomaterial which significantly promoted cementogenesis within the periodontal complex with structural integration by collagenous Sharpey's fiber insertions in vivo by controlling fibrin degradation and consequent cementoblast apoptosis. Furthermore, the modified fibrin could improve repair and regeneration of tooth roof-of-furcation defects, which has spatial curvatures and geometrical difficulties and hardly regenerates periodontal tissues.
Collapse
|
12
|
Zhang S, Kartha S, Lee J, Winkelstein BA. Techniques for Multiscale Neuronal Regulation via Therapeutic Materials and Drug Design. ACS Biomater Sci Eng 2017; 3:2744-2760. [DOI: 10.1021/acsbiomaterials.7b00012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Sijia Zhang
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich
Hall, Philadelphia, Pennsylvania 19104, United States
| | - Sonia Kartha
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich
Hall, Philadelphia, Pennsylvania 19104, United States
| | - Jasmine Lee
- Department of Physics and Astronomy, University of Pennsylvania, 209 S. 33rd Street, David Rittenhouse Laboratory, Philadelphia, Pennsylvania 19104, United States
| | - Beth A. Winkelstein
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich
Hall, Philadelphia, Pennsylvania 19104, United States
- Department
of Neurosurgery, University of Pennsylvania, Stemmler Hall, 3450 Hamilton Walk, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
13
|
CRISPR/Cas9-Mediated Genome Editing Corrects Dystrophin Mutation in Skeletal Muscle Stem Cells in a Mouse Model of Muscle Dystrophy. MOLECULAR THERAPY. NUCLEIC ACIDS 2017. [PMID: 28624206 PMCID: PMC5363682 DOI: 10.1016/j.omtn.2017.02.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Muscle stem cells (MuSCs) hold great therapeutic potential for muscle genetic disorders, such as Duchenne muscular dystrophy (DMD). The CRISP/Cas9-based genome editing is a promising technology for correcting genetic alterations in mutant genes. In this study, we used fibrin-gel culture system to selectively expand MuSCs from crude skeletal muscle cells of mdx mice, a mouse model of DMD. By CRISP/Cas9-based genome editing, we corrected the dystrophin mutation in expanded MuSCs and restored the skeletal muscle dystrophin expression upon transplantation in mdx mice. Our studies established a reliable and feasible platform for gene correction in MuSCs by genome editing, thus greatly advancing tissue stem cell-based therapies for DMD and other muscle disorders.
Collapse
|
14
|
Zhu P, Zhou Y, Wu F, Hong Y, Wang X, Shekhawat G, Mosenson J, Wu WS. Selective Expansion of Skeletal Muscle Stem Cells from Bulk Muscle Cells in Soft Three-Dimensional Fibrin Gel. Stem Cells Transl Med 2017; 6:1412-1423. [PMID: 28244269 PMCID: PMC5442710 DOI: 10.1002/sctm.16-0427] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/06/2017] [Indexed: 01/13/2023] Open
Abstract
Muscle stem cells (MuSCs) exhibit robust myogenic potential in vivo, thus providing a promising curative treatment for muscle disorders. Ex vivo expansion of adult MuSCs is highly desired to achieve a therapeutic cell dose because of their scarcity in limited muscle biopsies. Sorting of pure MuSCs is generally required for all the current culture systems. Here we developed a soft three‐dimensional (3D) salmon fibrin gel culture system that can selectively expand mouse MuSCs from bulk skeletal muscle preparations without cell sorting and faithfully maintain their regenerative capacity in culture. Our study established a novel platform for convenient ex vivo expansion of MuSCs, thus greatly advancing stem cell‐based therapies for various muscle disorders. Stem Cells Translational Medicine2017;6:1412–1423
Collapse
Affiliation(s)
- Pei Zhu
- Division of Hematology/Oncology, Department of Medicine and Cancer Center, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yalu Zhou
- Division of Hematology/Oncology, Department of Medicine and Cancer Center, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Furen Wu
- Division of Hematology/Oncology, Department of Medicine and Cancer Center, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yuanfan Hong
- Division of Hematology/Oncology, Department of Medicine and Cancer Center, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Xin Wang
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois, USA
| | - Gajendra Shekhawat
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois, USA
| | - Jeffrey Mosenson
- Division of Hematology/Oncology, Department of Medicine and Cancer Center, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Wen-Shu Wu
- Division of Hematology/Oncology, Department of Medicine and Cancer Center, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
15
|
Qiao S, Zhao Y, Geng S, Li Y, Hou X, Liu Y, Lin FH, Yao L, Tian W. A novel double-targeted nondrug delivery system for targeting cancer stem cells. Int J Nanomedicine 2016; 11:6667-6678. [PMID: 27994463 PMCID: PMC5154727 DOI: 10.2147/ijn.s116230] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Instead of killing cancer stem cells (CSCs), the conventional chemotherapy used for cancer treatment promotes the enrichment of CSCs, which are responsible for tumor growth, metastasis, and recurrence. However, most therapeutic agents are only able to kill a small proportion of CSCs by targeting one or two cell surface markers or dysregulated CSC pathways, which are usually shared with normal stem cells (NSCs). In this study, we developed a novel nondrug delivery system for the dual targeting of CSCs by conjugating hyaluronic acid (HA) and grafting the doublecortin-like kinase 1 (DCLK1) monoclonal antibody to the surface of poly(ethylene glycol) (PEG)–poly(d,l-lactide-co-glycolide) (PLGA) nanoparticles (NPs), which can specifically target CD44 receptors and the DCLK1 surface marker – the latter was shown to possess the capacity to distinguish between CSCSs and NSCs. The size and morphology of these NPs were characterized by dynamic light scattering (DLS), transmission electron microscopy (TEM), and scanning electron microscopy (SEM). This was followed by studies of NP encapsulation efficiency and in vitro drug release properties. Then, the cytotoxicity of the NPs was tested via Cell Counting Kit-8 assay. Finally, the 4T1 CSCs were obtained from the alginate-based platform, which we developed as an in vitro tumor model. Tumor-bearing nude mice were used as in vivo models to systematically detect the ability of NPs to target CSCs. Our results showed that the DCLK1–HA–PEG–PLGA NPs exhibited a targeting effect toward CSCs both in vitro and in vivo. These findings have important implications for the rational design of drug delivery systems that target CSCs with high efficacy.
Collapse
Affiliation(s)
- Shupei Qiao
- School of Life Science and Technology, Harbin Institute of Technology
| | - Yufang Zhao
- School of Life Science and Technology, Harbin Institute of Technology
| | - Shuai Geng
- Department of Pharmacology, Harbin Medical University
| | - Yong Li
- School of Life Science and Technology, Harbin Institute of Technology
| | - Xiaolu Hou
- School of Life Science and Technology, Harbin Institute of Technology; Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Yi Liu
- School of Life Science and Technology, Harbin Institute of Technology
| | - Feng-Huei Lin
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan; Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Lifen Yao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Weiming Tian
- School of Life Science and Technology, Harbin Institute of Technology
| |
Collapse
|
16
|
Arulmoli J, Wright HJ, Phan DTT, Sheth U, Que RA, Botten GA, Keating M, Botvinick EL, Pathak MM, Zarembinski TI, Yanni DS, Razorenova OV, Hughes CCW, Flanagan LA. Combination scaffolds of salmon fibrin, hyaluronic acid, and laminin for human neural stem cell and vascular tissue engineering. Acta Biomater 2016; 43:122-138. [PMID: 27475528 PMCID: PMC5386322 DOI: 10.1016/j.actbio.2016.07.043] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 06/29/2016] [Accepted: 07/26/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED Human neural stem/progenitor cells (hNSPCs) are good candidates for treating central nervous system (CNS) trauma since they secrete beneficial trophic factors and differentiate into mature CNS cells; however, many cells die after transplantation. This cell death can be ameliorated by inclusion of a biomaterial scaffold, making identification of optimal scaffolds for hNSPCs a critical research focus. We investigated the properties of fibrin-based scaffolds and their effects on hNSPCs and found that fibrin generated from salmon fibrinogen and thrombin stimulates greater hNSPC proliferation than mammalian fibrin. Fibrin scaffolds degrade over the course of a few days in vivo, so we sought to develop a novel scaffold that would retain the beneficial properties of fibrin but degrade more slowly to provide longer support for hNSPCs. We found combination scaffolds of salmon fibrin with interpenetrating networks (IPNs) of hyaluronic acid (HA) with and without laminin polymerize more effectively than fibrin alone and generate compliant hydrogels matching the physical properties of brain tissue. Furthermore, combination scaffolds support hNSPC proliferation and differentiation while significantly attenuating the cell-mediated degradation seen with fibrin alone. HNSPCs express two fibrinogen-binding integrins, αVβ1 and α5β1, and several laminin binding integrins (α7β1, α6β1, α3β1) that can mediate interaction with the scaffold. Lastly, to test the ability of scaffolds to support vascularization, we analyzed human cord blood-derived endothelial cells alone and in co-culture with hNSPCs and found enhanced vessel formation and complexity in co-cultures within combination scaffolds. Overall, combination scaffolds of fibrin, HA, and laminin are excellent biomaterials for hNSPCs. STATEMENT OF SIGNIFICANCE Interest has increased recently in the development of biomaterials as neural stem cell transplantation scaffolds to treat central nervous system (CNS) injury since scaffolds improve survival and integration of transplanted cells. We report here on a novel combination scaffold composed of fibrin, hyaluronic acid, and laminin to support human neural stem/progenitor cell (hNSPC) function. This combined biomaterial scaffold has appropriate physical properties for hNSPCs and the CNS, supports hNSPC proliferation and differentiation, and attenuates rapid cell-mediated scaffold degradation. The hNSPCs and scaffold components synergistically encourage new vessel formation from human endothelial cells. This work marks the first report of a combination scaffold supporting human neural and vascular cells to encourage vasculogenesis, and sets a benchmark for biomaterials to treat CNS injury.
Collapse
Affiliation(s)
- Janahan Arulmoli
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Heather J Wright
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Duc T T Phan
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Urmi Sheth
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Richard A Que
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Giovanni A Botten
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mark Keating
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Elliot L Botvinick
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA
| | - Medha M Pathak
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | | | - Daniel S Yanni
- Disc Comfort, Inc., 351 Hospital Road, Suite 202, Newport Beach, CA 92663, USA
| | - Olga V Razorenova
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Christopher C W Hughes
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA
| | - Lisa A Flanagan
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
17
|
Urbanski MM, Kingsbury L, Moussouros D, Kassim I, Mehjabeen S, Paknejad N, Melendez-Vasquez CV. Myelinating glia differentiation is regulated by extracellular matrix elasticity. Sci Rep 2016; 6:33751. [PMID: 27646171 PMCID: PMC5028715 DOI: 10.1038/srep33751] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/02/2016] [Indexed: 12/27/2022] Open
Abstract
The mechanical properties of living tissues have a significant impact on cell differentiation, but remain unexplored in the context of myelin formation and repair. In the PNS, the extracellular matrix (ECM) incorporates a basal lamina significantly denser than the loosely organized CNS matrix. Inhibition of non-muscle myosin II (NMII) enhances central but impairs peripheral myelination and NMII has been implicated in cellular responses to changes in the elasticity of the ECM. To directly evaluate whether mechanotransduction plays a role in glial cell differentiation, we cultured Schwann cells (SC) and oligodendrocytes (OL) on matrices of variable elastic modulus, mimicking either their native environment or conditions found in injured tissue. We found that a rigid, lesion-like matrix inhibited branching and differentiation of OL in NMII-dependent manner. By contrast, SC developed normally in both soft and stiffer matrices. Although SC differentiation was not significantly affected by changes in matrix stiffness alone, we found that expression of Krox-20 was potentiated on rigid matrices at high laminin concentration. These findings are relevant to the design of biomaterials to promote healing and regeneration in both CNS and PNS, via transplantation of glial progenitors or the implantation of tissue scaffolds.
Collapse
Affiliation(s)
- Mateusz M Urbanski
- Hunter College, Department of Biological Sciences, New York, NY 10065, USA.,The Graduate Center, Molecular Cellular and Developmental Biology, The City University of New York, NY 10016, USA
| | - Lyle Kingsbury
- Hunter College, Department of Biological Sciences, New York, NY 10065, USA
| | - Daniel Moussouros
- Hunter College, Department of Biological Sciences, New York, NY 10065, USA
| | - Imran Kassim
- Hunter College, Department of Biological Sciences, New York, NY 10065, USA
| | - Saraf Mehjabeen
- Hunter College, Department of Biological Sciences, New York, NY 10065, USA
| | - Navid Paknejad
- Molecular Cytology Core Facility, Zuckerman Research Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carmen V Melendez-Vasquez
- Hunter College, Department of Biological Sciences, New York, NY 10065, USA.,The Graduate Center, Molecular Cellular and Developmental Biology, The City University of New York, NY 10016, USA
| |
Collapse
|
18
|
Smith JR, Galie PA, Slochower DR, Weisshaar CL, Janmey PA, Winkelstein BA. Salmon-derived thrombin inhibits development of chronic pain through an endothelial barrier protective mechanism dependent on APC. Biomaterials 2015; 80:96-105. [PMID: 26708087 DOI: 10.1016/j.biomaterials.2015.11.062] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/13/2015] [Accepted: 11/29/2015] [Indexed: 02/08/2023]
Abstract
Many neurological disorders are initiated by blood-brain barrier breakdown, which potentiates spinal neuroinflammation and neurodegeneration. Peripheral neuropathic injuries are known to disrupt the blood-spinal cord barrier (BSCB) and to potentiate inflammation. But, it is not known whether BSCB breakdown facilitates pain development. In this study, a neural compression model in the rat was used to evaluate relationships among BSCB permeability, inflammation and pain-related behaviors. BSCB permeability increases transiently only after injury that induces mechanical hyperalgesia, which correlates with serum concentrations of pro-inflammatory cytokines, IL-7, IL-12, IL-1α and TNF-α. Mammalian thrombin dually regulates vascular permeability through PAR1 and activated protein C (APC). Since thrombin protects vascular integrity through APC, directing its affinity towards protein C, while still promoting coagulation, might be an ideal treatment for BSCB-disrupting disorders. Salmon thrombin, which prevents the development of mechanical allodynia, also prevents BSCB breakdown after neural injury and actively inhibits TNF-α-induced endothelial permeability in vitro, which is not evident the case for human thrombin. Salmon thrombin's production of APC faster than human thrombin is confirmed using a fluorogenic assay and APC is shown to inhibit BSCB breakdown and pain-related behaviors similar to salmon thrombin. Together, these studies highlight the impact of BSCB on pain and establish salmon thrombin as an effective blocker of BSCB, and resulting nociception, through its preferential affinity for protein C.
Collapse
Affiliation(s)
- Jenell R Smith
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter A Galie
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David R Slochower
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christine L Weisshaar
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul A Janmey
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Beth A Winkelstein
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
19
|
AAVshRNA-mediated suppression of PTEN in adult rats in combination with salmon fibrin administration enables regenerative growth of corticospinal axons and enhances recovery of voluntary motor function after cervical spinal cord injury. J Neurosci 2014; 34:9951-62. [PMID: 25057197 DOI: 10.1523/jneurosci.1996-14.2014] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Conditional genetic deletion of phosphatase and tensin homolog (PTEN) in the sensorimotor cortex of neonatal mice enables regeneration of corticospinal tract (CST) axons after spinal cord injury (SCI). The present study addresses three questions: (1) whether PTEN knockdown in adult rats by nongenetic techniques enables CST regeneration, (2) whether interventions to enable CST regeneration enhance recovery of voluntary motor function, and (3) whether delivery of salmon fibrin into the injury site further enhances CST regeneration and motor recovery. Adult rats were trained in a staircase-reaching task and then received either intracortical injections of AAVshPTEN to delete PTEN or a control vector expressing shRNA for luciferase (AAVshLuc). Rats then received cervical dorsal hemisection injuries and salmon fibrin was injected into the injury site in half the rats, yielding four groups (AAVshPTEN, AAVshLuc, AAVshPTEN + fibrin, and AAVshLuc + fibrin). Forepaw function was assessed for 10 weeks after injury and CST axons were traced by injecting biotin-conjugated dextran amine into the sensorimotor cortex. Rats that received AAVshPTEN alone did not exhibit improved motor function, whereas rats that received AAVshPTEN and salmon fibrin had significantly higher forelimb-reaching scores. Tract tracing revealed that CST axons extended farther caudally in the group that received AAVshPTEN and salmon fibrin versus other groups. There were no significant differences in lesion size between the groups. Together, these data suggest that the combination of PTEN deletion and salmon fibrin injection into the lesion can significantly improve voluntary motor function after SCI by enabling regenerative growth of CST axons.
Collapse
|
20
|
Salmon and human thrombin differentially regulate radicular pain, glial-induced inflammation and spinal neuronal excitability through protease-activated receptor-1. PLoS One 2013; 8:e80006. [PMID: 24278231 PMCID: PMC3835785 DOI: 10.1371/journal.pone.0080006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/07/2013] [Indexed: 11/22/2022] Open
Abstract
Chronic neck pain is a major problem with common causes including disc herniation and spondylosis that compress the spinal nerve roots. Cervical nerve root compression in the rat produces sustained behavioral hypersensitivity, due in part to the early upregulation of pro-inflammatory cytokines, the sustained hyperexcitability of neurons in the spinal cord and degeneration in the injured nerve root. Through its activation of the protease-activated receptor-1 (PAR1), mammalian thrombin can enhance pain and inflammation; yet at lower concentrations it is also capable of transiently attenuating pain which suggests that PAR1 activation rate may affect pain maintenance. Interestingly, salmon-derived fibrin, which contains salmon thrombin, attenuates nerve root-induced pain and inflammation, but the mechanisms of action leading to its analgesia are unknown. This study evaluates the effects of salmon thrombin on nerve root-mediated pain, axonal degeneration in the root, spinal neuronal hyperexcitability and inflammation compared to its human counterpart in the context of their enzymatic capabilities towards coagulation substrates and PAR1. Salmon thrombin significantly reduces behavioral sensitivity, preserves neuronal myelination, reduces macrophage infiltration in the injured nerve root and significantly decreases spinal neuronal hyperexcitability after painful root compression in the rat; whereas human thrombin has no effect. Unlike salmon thrombin, human thrombin upregulates the transcription of IL-1β and TNF-α and the secretion of IL-6 by cortical cultures. Salmon and human thrombins cleave human fibrinogen-derived peptides and form clots with fibrinogen with similar enzymatic activities, but salmon thrombin retains a higher enzymatic activity towards coagulation substrates in the presence of antithrombin III and hirudin compared to human thrombin. Conversely, salmon thrombin activates a PAR1-derived peptide more weakly than human thrombin. These results are the first to demonstrate that salmon thrombin has unique analgesic, neuroprotective and anti-inflammatory capabilities compared to human thrombin and that PAR1 may contribute to these actions.
Collapse
|
21
|
Asmani MN, Ai J, Amoabediny G, Noroozi A, Azami M, Ebrahimi-Barough S, Navaei-Nigjeh M, Ai A, Jafarabadi M. Three-dimensional culture of differentiated endometrial stromal cells to oligodendrocyte progenitor cells (OPCs) in fibrin hydrogel. Cell Biol Int 2013; 37:1340-9. [PMID: 24038753 DOI: 10.1002/cbin.10171] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Accepted: 07/22/2012] [Indexed: 01/08/2023]
Abstract
Neural tissue engineering is one of the most promising strategies for treatment of nerve tissue injuries. Three-dimensional (3D) environment mimics in vivo conditions for cells. 3D distribution and growth of the cells within the scaffold are both important for neural tissue engineering. In this study, endometrial stromal cell-derived oligodendrocyte progenitor cells (EnSC-derived OPCs) were cultured in fibrin gel and cell differentiation and viability were evaluated after 8 days of post-culture. The structural and mechanical characteristics of fibrin gel-like scaffold were examined with rheological analysis. EnSCs were isolated from donor tissue and were induced to OPCs with growth factors (FGF2/EGF/PDGF-AA) for 12 days, then were cultured in fibrin gel with Triiodothyronine (T3) medium for another 8 days. The viability of cells was analyzed using MTT assay for a period of 8 days culturing in a fibrin matrix. Structure of fibrin matrix and cell morphology was analyzed with SEM. TEM, immunostaining and quantitative RT-PCR was performed for OPCs markers after cell culturing in fibrin matrix. Cell viability is enhanced in fibrin matrix after 8 days. SEM and TEM show that cells are in good integration with nano-fibers. Moreover, immunohistochemistry and quantitative RT-PCR of OPCs differentiation markers showed that Olig2, Sox10, PDGFRa, CNP, and A2B5 are expressed after 8 days culturing within fibrin matrix. Fibrin can provide a suitable 3-D scaffold for EnSCs differentiated cells for the regeneration of CNS.
Collapse
Affiliation(s)
- Mohammad Nabi Asmani
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran; Faculty of New Sciences & Technologies, University of Tehran, Tehran, Iran
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Vetrik M, Pradny M, Kobera L, Slouf M, Rabyk M, Pospisilova A, Stepanek P, Hruby M. Biopolymer-based degradable nanofibres from renewable resources produced by freeze-drying. RSC Adv 2013. [DOI: 10.1039/c3ra42647e] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
23
|
Liu J, Tan Y, Zhang H, Zhang Y, Xu P, Chen J, Poh YC, Tang K, Wang N, Huang B. Soft fibrin gels promote selection and growth of tumorigenic cells. NATURE MATERIALS 2012; 11:734-41. [PMID: 22751180 PMCID: PMC3405191 DOI: 10.1038/nmat3361] [Citation(s) in RCA: 347] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 05/17/2012] [Indexed: 05/15/2023]
Abstract
The identification of stem-cell-like cancer cells through conventional methods that depend on stem cell markers is often unreliable. We developed a mechanical method for selecting tumorigenic cells by culturing single cancer cells in fibrin matrices of ~100 Pa in stiffness. When cultured within these gels, primary human cancer cells or single cancer cells from mouse or human cancer cell lines grew within a few days into individual round colonies that resembled embryonic stem cell colonies. Subcutaneous or intravenous injection of 10 or 100 fibrin-cultured cells in syngeneic or severe combined immunodeficiency mice led to the formation of solid tumours at the site of injection or at the distant lung organ much more efficiently than control cancer cells selected using conventional surface marker methods or cultured on conventional rigid dishes or on soft gels. Remarkably, as few as ten such cells were able to survive and form tumours in the lungs of wild-type non-syngeneic mice.
Collapse
Affiliation(s)
- Jing Liu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030 China
| | - Youhua Tan
- Department of Mechanical Science and Engineering, College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Huafeng Zhang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030 China
| | - Yi Zhang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030 China
| | - Pingwei Xu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030 China
| | - Junwei Chen
- Laboratory for Cell Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074 China
| | - Yeh-Chuin Poh
- Department of Mechanical Science and Engineering, College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Ke Tang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030 China
| | - Ning Wang
- Laboratory for Cell Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074 China
- Department of Mechanical Science and Engineering, College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
- Correspondence should be addressed to: Ning Wang () or Bo Huang ()
| | - Bo Huang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030 China
- Department of Immunology, Institute of Basic Medical Sciences of Chinese Academy of Medical Sciences, Beijing 100005 China
- Correspondence should be addressed to: Ning Wang () or Bo Huang ()
| |
Collapse
|
24
|
Advances in natural biomaterials for nerve tissue repair. Neurosci Lett 2012; 519:103-14. [DOI: 10.1016/j.neulet.2012.02.027] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 02/06/2012] [Accepted: 02/08/2012] [Indexed: 12/22/2022]
|
25
|
Keung AJ, de Juan-Pardo EM, Schaffer DV, Kumar S. Rho GTPases mediate the mechanosensitive lineage commitment of neural stem cells. Stem Cells 2012; 29:1886-97. [PMID: 21956892 DOI: 10.1002/stem.746] [Citation(s) in RCA: 164] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Adult neural stem cells (NSCs) play important roles in learning and memory and are negatively impacted by neurological disease. It is known that biochemical and genetic factors regulate self-renewal and differentiation, and it has recently been suggested that mechanical and solid-state cues, such as extracellular matrix (ECM) stiffness, can also regulate the functions of NSCs and other stem cell types. However, relatively little is known of the molecular mechanisms through which stem cells transduce mechanical inputs into fate decisions, the extent to which mechanical inputs instruct fate decisions versus select for or against lineage-committed blast populations, or the in vivo relevance of mechanotransductive signaling molecules in native stem cell niches. Here we demonstrate that ECM-derived mechanical signals act through Rho GTPases to activate the cellular contractility machinery in a key early window during differentiation to regulate NSC lineage commitment. Furthermore, culturing NSCs on increasingly stiff ECMs enhances RhoA and Cdc42 activation, increases NSC stiffness, and suppresses neurogenesis. Likewise, inhibiting RhoA and Cdc42 or downstream regulators of cellular contractility rescues NSCs from stiff matrix- and Rho GTPase-induced neurosuppression. Importantly, Rho GTPase expression and ECM stiffness do not alter proliferation or apoptosis rates indicating that an instructive rather than selective mechanism modulates lineage distributions. Finally, in the adult brain, RhoA activation in hippocampal progenitors suppresses neurogenesis, analogous to its effect in vitro. These results establish Rho GTPase-based mechanotransduction and cellular stiffness as biophysical regulators of NSC fate in vitro and RhoA as an important regulatory protein in the hippocampal stem cell niche.
Collapse
Affiliation(s)
- Albert J Keung
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California 94720-3220, USA
| | | | | | | |
Collapse
|
26
|
Ziv-Polat O, Skaat H, Shahar A, Margel S. Novel magnetic fibrin hydrogel scaffolds containing thrombin and growth factors conjugated iron oxide nanoparticles for tissue engineering. Int J Nanomedicine 2012; 7:1259-74. [PMID: 22419873 PMCID: PMC3298389 DOI: 10.2147/ijn.s26533] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Novel tissue-engineered magnetic fibrin hydrogel scaffolds were prepared by the interaction of thrombin-conjugated iron oxide magnetic nanoparticles with fibrinogen. In addition, stabilization of basal fibroblast growth factor (bFGF) was achieved by the covalent and physical conjugation of the growth factor to the magnetic nanoparticles. Adult nasal olfactory mucosa (NOM) cells were seeded in the transparent fibrin scaffolds in the absence or presence of the free or conjugated bFGF-iron oxide nanoparticles. The conjugated bFGF enhanced significantly the growth and differentiation of the NOM cells in the fibrin scaffolds, compared to the same or even five times higher concentration of the free bFGF. In the presence of the bFGF-conjugated magnetic nanoparticles, the cultured NOM cells proliferated and formed a three-dimensional interconnected network composed mainly of tapered bipolar cells. The magnetic properties of these matrices are due to the integration of the thrombin- and bFGF-conjugated magnetic nanoparticles within the scaffolds. The magnetic properties of these scaffolds may be used in future work for various applications, such as magnetic resonance visualization of the scaffolds after implantation and reloading the scaffolds via magnetic forces with bioactive agents, eg, growth factors bound to the iron oxide magnetic nanoparticles.
Collapse
Affiliation(s)
- Ofra Ziv-Polat
- Department of Chemistry, Bar-Ilan Institute of Nanotechnology and Advanced Materials, Ramat-Gan, Israel
| | | | | | | |
Collapse
|
27
|
Sharp KG, Dickson AR, Marchenko SA, Yee KM, Emery PN, Laidmåe I, Uibo R, Sawyer ES, Steward O, Flanagan LA. Salmon fibrin treatment of spinal cord injury promotes functional recovery and density of serotonergic innervation. Exp Neurol 2012; 235:345-56. [PMID: 22414309 DOI: 10.1016/j.expneurol.2012.02.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 01/25/2012] [Accepted: 02/25/2012] [Indexed: 12/19/2022]
Abstract
The neural degeneration caused by spinal cord injury leaves a cavity at the injury site that greatly inhibits repair. One approach to promoting repair is to fill the cavity with a scaffold to limit further damage and encourage regrowth. Injectable materials are advantageous scaffolds because they can be placed as a liquid in the lesion site then form a solid in vivo that precisely matches the contours of the lesion. Fibrin is one type of injectable scaffold, but risk of infection from blood borne pathogens has limited its use. We investigated the potential utility of salmon fibrin as an injectable scaffold to treat spinal cord injury since it lacks mammalian infectious agents and encourages greater neuronal extension in vitro than mammalian fibrin or Matrigel®, another injectable material. Female rats received a T9 dorsal hemisection injury and were treated with either salmon or human fibrin at the time of injury while a third group served as untreated controls. Locomotor function was assessed using the BBB scale, bladder function was analyzed by measuring residual urine, and sensory responses were tested by mechanical stimulation (von Frey hairs). Histological analyses quantified the glial scar, lesion volume, and serotonergic fiber density. Rats that received salmon fibrin exhibited significantly improved recovery of both locomotor and bladder function and a greater density of serotonergic innervation caudal to the lesion site without exacerbation of pain. Rats treated with salmon fibrin also exhibited less autophagia than those treated with human fibrin, potentially pointing to amelioration of sensory dysfunction. Glial scar formation and lesion size did not differ significantly among groups. The pattern and timing of salmon fibrin's effects suggest that it acts on neuronal populations but not by stimulating long tract regeneration. Salmon fibrin clearly has properties distinct from those of mammalian fibrin and is a beneficial injectable scaffold for treatment of spinal cord injury.
Collapse
Affiliation(s)
- Kelli G Sharp
- Reeve-Irvine Research Center and Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA 92697-1705, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Spinal cord injury (SCI) presents a complex regenerative problem due to the multiple facets of growth inhibition that occur following trauma to the cord parenchyma and stroma. Clinically, SCI is further complicated by the heterogeneity in the size, shape and extent of human injuries. Many of these injuries do not breach the dura mater and have continuous viable axons through the injury site that can later lead to some degree of functional recovery. In these cases, surgical manipulation of the spinal cord by implanting a preformed scaffold or drug delivery device may lead to further damage. Given these circumstances, in situ-forming scaffolds are an attractive approach for SCI regeneration. These synthetic and natural polymers undergo a rapid transformation from liquid to gel upon injection into the cord tissue, conforming to the individual lesion site and directly integrating with the host tissue. Injectable materials can be formulated to have mechanical properties that closely match the native spinal cord extracellular matrix, and this may enhance axonal ingrowth. Such materials can also be loaded with cellular and molecular therapeutics to modulate the wound environment and enhance regeneration. This review will focus on the current status of in situ-forming materials for spinal cord repair. The advantages of, and requirements for, such polymers will be presented, and examples of the behavior of such systems in vitro and in vivo will be presented. There are helpful lessons to be learned from the investigations of injectable hydrogels for the treatment of SCI that apply to the use of these biomaterials for the treatment of lesions in other central nervous system tissues and in organs comprising other tissue types.
Collapse
Affiliation(s)
- D Macaya
- Tissue Engineering, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
29
|
Barsotti MC, Felice F, Balbarini A, Di Stefano R. Fibrin as a scaffold for cardiac tissue engineering. Biotechnol Appl Biochem 2011; 58:301-10. [PMID: 21995533 DOI: 10.1002/bab.49] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 08/12/2011] [Indexed: 12/16/2022]
Abstract
Fibrin is a natural biopolymer with many interesting properties, such as biocompatibility, bioresorbability, ease of processing, ability to be tailored to modify the conditions of polymerization, and potential for incorporation of both cells and cell mediators. Moreover, the fibrin network has a nanometric fibrous structure, mimicking extracellular matrix, and it can also be used in autologous applications. Therefore, fibrin has found many applications in tissue engineering, combined with cells, growth factors, or drugs. Because a major limitation of cardiac cell therapy is low cell engraftment, the use of biodegradable scaffolds for specific homing and in situ cell retention is desirable. Thus, fibrin-based injectable cardiac tissue engineering may enhance cell therapy efficacy. Fibrin-based biomaterials can also be used for engineering heart valves or cardiac patches. The aim of this review is to show cardiac bioengineering uses of fibrin, both as a cell delivery vehicle and as an implantable biomaterial.
Collapse
Affiliation(s)
- Maria Chiara Barsotti
- Cardiovascular Research Laboratory, Cardiac, Thoracic and Vascular Department, University of Pisa, Pisa, Italy.
| | | | | | | |
Collapse
|
30
|
Weisshaar CL, Winer JP, Guarino BB, Janmey PA, Winkelstein BA. The potential for salmon fibrin and thrombin to mitigate pain subsequent to cervical nerve root injury. Biomaterials 2011; 32:9738-46. [PMID: 21944723 DOI: 10.1016/j.biomaterials.2011.09.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 09/07/2011] [Indexed: 01/23/2023]
Abstract
Nerve root compression is a common cause of radiculopathy and induces persistent pain. Mammalian fibrin is used clinically as a coagulant but presents a variety of risks. Fish fibrin is a potential biomaterial for neural injury treatment because it promotes neurite outgrowth, is non-toxic, and clots readily at lower temperatures. This study administered salmon fibrin and thrombin following nerve root compression and measured behavioral sensitivity and glial activation in a rat pain model. Fibrin and thrombin each significantly reduced mechanical allodynia compared to injury alone (p < 0.02). Painful compression with fibrin exhibited allodynia that was not different from sham for any day using stimulation by a 2 g filament; allodynia was only significantly different (p < 0.043) from sham using the 4 g filament on days 1 and 3. By day 5, responses for fibrin treatment decreased to sham levels. Allodynia following compression with thrombin treatment were unchanged from sham at any time point. Macrophage infiltration at the nerve root and spinal microglial activation were only mildly modified by salmon treatments. Spinal astrocytic expression decreased significantly with fibrin (p < 0.0001) but was unchanged from injury responses for thrombin treatment. Results suggest that salmon fibrin and thrombin may be suitable biomaterials to mitigate pain.
Collapse
Affiliation(s)
- Christine L Weisshaar
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104-6321, USA.
| | | | | | | | | |
Collapse
|
31
|
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
|
32
|
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]
|
33
|
Bandiera A, Sist P, Urbani R. Comparison of Thermal Behavior of Two Recombinantly Expressed Human Elastin-Like Polypeptides for Cell Culture Applications. Biomacromolecules 2010; 11:3256-65. [DOI: 10.1021/bm100644m] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Antonella Bandiera
- Department of Life Sciences, University of Trieste, via L. Giorgieri, 1, 34127 Trieste, Italy
| | - Paola Sist
- Department of Life Sciences, University of Trieste, via L. Giorgieri, 1, 34127 Trieste, Italy
| | - Ranieri Urbani
- Department of Life Sciences, University of Trieste, via L. Giorgieri, 1, 34127 Trieste, Italy
| |
Collapse
|