Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biomacromolecules ; 21(2): 276-293, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31789020

ABSTRACT

Laminin is a heterotrimeric glycoprotein with a key role in the formation and maintenance of the basement membrane architecture and properties, as well as on the modulation of several biological functions, including cell adhesion, migration, differentiation and matrix-mediated signaling. In the central nervous system (CNS), laminin is differentially expressed during development and homeostasis, with an impact on the modulation of cell function and fate. Within neurogenic niches, laminin is one of the most important and well described extracellular matrix (ECM) proteins. Specifically, efforts have been made to understand laminin assembly, domain architecture, and interaction of its different bioactive domains with cell surface receptors, soluble signaling molecules, and ECM proteins, to gain insight into the role of this ECM protein and its receptors on the modulation of neurogenesis, both in homeostasis and during repair. This is also expected to provide a rational basis for the design of biomaterial-based matrices mirroring the biological properties of the basement membrane of neural stem cell niches, for application in neural tissue repair and cell transplantation. This review provides a general overview of laminin structure and domain architecture, as well as the main biological functions mediated by this heterotrimeric glycoprotein. The expression and distribution of laminin in the CNS and, more specifically, its role within adult neural stem cell niches is summarized. Additionally, a detailed overview on the use of full-length laminin and laminin derived peptide/recombinant laminin fragments for the development of hydrogels for mimicking the neurogenic niche microenvironment is given. Finally, the main challenges associated with the development of laminin-inspired hydrogels and the hurdles to overcome for these to progress from bench to bedside are discussed.


Subject(s)
Central Nervous System/metabolism , Hydrogels/chemistry , Laminin/chemistry , Laminin/physiology , Neural Stem Cells/cytology , Animals , Central Nervous System/cytology , Humans , Neural Stem Cells/physiology , Peptides/chemistry
2.
Biomater Sci ; 7(12): 5338-5349, 2019 Nov 19.
Article in English | MEDLINE | ID: mdl-31620727

ABSTRACT

Laminin incorporation into biological or synthetic hydrogels has been explored to recapitulate the dynamic nature and biological complexity of neural stem cell (NSC) niches. However, the strategies currently explored for laminin immobilization within three-dimensional (3D) matrices do not address a critical aspect influencing cell-matrix interactions, which is the control over laminin conformation and orientation upon immobilization. This is a key feature for the preservation of the protein bioactivity. In this work, we explored an affinity-based approach to mediate the site-selective immobilization of laminin into a degradable synthetic hydrogel. Specifically, a four-arm maleimide terminated poly(ethylene glycol) (PEG-4MAL) macromer was functionalized with a mono-PEGylated recombinant human N-terminal agrin (NtA) domain, to promote high affinity binding of laminin. Different NtA concentrations (10, 50 and 100 µM) were used to investigate the impact of NtA density on laminin incorporation, hydrogel biophysical properties, and biological outcome. Laminin was efficiently incorporated for all the conditions tested (laminin incorporation >95%), and the developed hydrogels revealed mechanical properties (average storage modulus (G') ranging from 187 to 256 Pa) within the values preferred for NSC proliferation and neurite branching and extension. Affinity-bound laminin PEG-4MAL hydrogels better preserve laminin bioactivity, compared to unmodified hydrogels and hydrogels containing physically entrapped laminin, this effect being dependent on NtA concentration. This was evidenced by the 10 µM NtA-functionalized PEG-4MAL gels incorporating laminin that support enhanced human NSC proliferation and neurite extension, compared to the latter. Overall, this work highlights the potential of the proposed engineered matrices to be used as defined 3D platforms for the establishment of artificial NSC niches and as extracellular matrix-mimetic microenvironments to support human NSC transplantation.


Subject(s)
Engineering , Hydrogels/chemistry , Hydrogels/pharmacology , Laminin/chemistry , Maleimides/chemistry , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Neuronal Outgrowth/drug effects , Neurons/cytology , Neurons/drug effects , Phenotype , Polyethylene Glycols/chemistry
3.
Methods Mol Biol ; 2036: 205-219, 2019.
Article in English | MEDLINE | ID: mdl-31410799

ABSTRACT

Biomaterials-based hydrogels are attractive drug-eluting vehicles in the context of RNA therapeutics, such as those utilizing antisense oligonucleotide or RNA interference based drugs, as they can potentially reduce systemic toxicity and enhance in vivo efficacy by increasing in situ concentrations. Here we describe the preparation of antisense oligonucleotide-loaded fibrin hydrogels exploring their applications in the context of the nervous system utilizing an organotypic dorsal root ganglion explant in vitro system and an in vivo model of spinal cord injury.


Subject(s)
Drug Carriers , Hydrogels/chemistry , Oligonucleotides, Antisense/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems , Fibrin/chemistry , Ganglia, Spinal/metabolism , Gene Silencing , Humans , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/genetics , Spinal Cord
4.
Mol Ther Nucleic Acids ; 11: 393-406, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29858074

ABSTRACT

After spinal cord injury (SCI), nerve regeneration is severely hampered due to the establishment of a highly inhibitory microenvironment at the injury site, through the contribution of multiple factors. The potential of antisense oligonucleotides (AONs) to modify gene expression at different levels, allowing the regulation of cell survival and cell function, together with the availability of chemically modified nucleic acids with favorable biopharmaceutical properties, make AONs an attractive tool for novel SCI therapy developments. In this work, we explored the potential of locked nucleic acid (LNA)-modified AON gapmers in combination with a fibrin hydrogel bridging material to induce gene silencing in situ at a SCI lesion site. LNA gapmers were effectively developed against two promising gene targets aiming at enhancing axonal regeneration-RhoA and GSK3ß. The fibrin-matrix-assisted AON delivery system mediated potent RNA knockdown in vitro in a dorsal root ganglion explant culture system and in vivo at a SCI lesion site, achieving around 75% downregulation 5 days after hydrogel injection. Our results show that local implantation of a AON-gapmer-loaded hydrogel matrix mediated efficient gene silencing in the lesioned spinal cord and is an innovative platform that can potentially combine gene regulation with regenerative permissive substrates aiming at SCI therapeutics and nerve regeneration.

5.
Acta Biomater ; 59: 243-256, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28694236

ABSTRACT

To enhance fibrin hydrogel affinity towards pluripotent stem cell-derived neural stem/progenitor cells (NSPCs) and its capacity to support NSPC migration and neurite extension, we explored the tethering of synthetic peptides engaging integrin α6ß1, a cell receptor enriched in NSPCs. Six α6ß1 integrin ligands were tested for their ability to support integrin α6ß1-mediated adhesion of embryonic stem cell-derived NSPCs (ES-NSPs) and sustain ES-NSPC viability, migration, and neuronal differentiation. Due to their better performance, peptides T1, HYD1, and A5G81 were immobilized into fibrin and functionalized gels characterized in terms of peptide binding efficiency, structure and viscoelastic properties. Tethering of T1 or HYD1 successfully enhanced cell outgrowth from ES-NSPC neurospheres (up to 2.4-fold increase), which exhibited a biphasic response to peptide concentration. Inhibition assays evidenced the involvement of α6ß1 and α3ß1 integrins in mediating radial outgrowth on T1-/HYD1-functionalized gels. Fibrin functionalization also promoted neurite extension of single ES-NSPCs in fibrin, without affecting cell proliferation and neuronal differentiation. Finally, HYD1-functionalized gels were found to provide a permissive environment for axonal regeneration, leading up to a 2.0-fold increase in neurite extension from rat dorsal root ganglia explants as compared to unmodified fibrin, and to significant improved locomotor function after spinal cord injury (complete transection), along with a trend toward a higher area positive for growth associated protein 43 (marker for axonal growth cone formation). Our results suggest that conjugation of α6ß1 integrin-binding motifs is of interest to increase the biofunctionality of hydrogels used in 3D platforms for ES-NSPC culture and potentially, in matrix-assisted ES-NSPC transplantation. STATEMENT OF SIGNIFICANCE: Impact statement: The transplantation of NSPCs derived from pluripotent stem cells holds much promise for the treatment of central nervous system disorders. Moreover, the combinatorial use of biodegradable hydrogels with NSPCs was shown to contribute to the establishment of a more permissive environment for survival and integration of transplanted cells. In this study, fibrin hydrogels functionalized with a synthetic peptide engaging integrin α6ß1 (HYD1) were shown to promote neurite extension of ES-NSPCs, which is fundamental for the formation of functional neuronal relay circuits after NSPC transplantation. Notably, HYD1-functionalized fibrin per se led to enhanced axonal growth ex vivo and to an improvement in locomotor function after implantation in a rat model of spinal cord injury. Conjugation of α6ß1 integrin-binding motifs may therefore be of interest to confer bioactivity to NSPC hydrogel vehicles.


Subject(s)
Embryonic Stem Cells/metabolism , Fibrin/chemistry , Integrin alpha6beta1/metabolism , Neural Stem Cells/metabolism , Neurites/metabolism , Animals , Cell Line, Tumor , Embryonic Stem Cells/cytology , Humans , Ligands , Mice , Neural Stem Cells/cytology , Rats , Rats, Wistar
6.
J Tissue Eng Regen Med ; 11(8): 2227-2240, 2017 08.
Article in English | MEDLINE | ID: mdl-26880706

ABSTRACT

Embryonic stem (ES)-derived neural stem/progenitor cells (ES-NSPCs) constitute a promising cell source for application in cell therapies for the treatment of central nervous system disorders. In this study, a rotary orbital hydrodynamic culture system was applied to single-cell suspensions of ES-NSPCs, to obtain homogeneously-sized ES-NSPC cellular aggregates (neurospheres). Hydrodynamic culture allowed the formation of ES-NSPC neurospheres with a narrower size distribution than statically cultured neurospheres, increasing orbital speeds leading to smaller-sized neurospheres and higher neurosphere yield. Neurospheres formed under hydrodynamic conditions (72 h at 55 rpm) showed higher cell compaction and comparable percentages of viable, dead, apoptotic and proliferative cells. Further characterization of cellular aggregates provided new insights into the effect of hydrodynamic shear on ES-NSPC behaviour. Rotary neurospheres exhibited reduced protein levels of N-cadherin and ß-catenin, and higher deposition of laminin (without impacting fibronectin deposition), matrix metalloproteinase-2 (MMP-2) activity and percentage of neuronal cells. In line with the increased MMP-2 activity levels found, hydrodynamically-cultured neurospheres showed higher outward migration on laminin. Moreover, when cultured in a 3D fibrin hydrogel, rotary neurospheres generated an increased percentage of neuronal cells. In conclusion, the application of a constant orbital speed to single-cell suspensions of ES-NSPCs, besides allowing the formation of homogeneously-sized neurospheres, promoted ES-NSPC differentiation and outward migration, possibly by influencing the expression of cell-cell adhesion molecules and the secretion of proteases/extracellular matrix proteins. These findings are important when establishing the culture conditions needed to obtain uniformly-sized ES-NSPC aggregates, either for use in regenerative therapies or in in vitro platforms for biomaterial development or pharmacological screening. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Cell Culture Techniques/methods , Hydrodynamics , Mouse Embryonic Stem Cells , Neural Stem Cells , Spheroids, Cellular , Animals , Cell Culture Techniques/instrumentation , Cell Line , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism
7.
J Tissue Eng Regen Med ; 11(12): 3494-3507, 2017 12.
Article in English | MEDLINE | ID: mdl-28032468

ABSTRACT

In an attempt to improve the efficacy of neural stem/progenitor cell (NSPC) based therapies, fibrin hydrogels are being explored to provide a favourable microenvironment for cell survival and differentiation following transplantation. In the present work, the ability of fibrin to support the survival, proliferation, and neuronal differentiation of NSPCs derived from embryonic stem (ES) cells under monolayer culture was explored. Single mouse ES-NSPCs were cultured within fibrin (fibrinogen concentration: 6 mg/ml) under neuronal differentiation conditions up to 14 days. The ES-NSPCs retained high cell viability and proliferated within small-sized spheroids. Neuronal differentiation was confirmed by an increase in the levels of ßIII-tubulin and NF200 over time. At day 14, cell-matrix constructs mainly comprised NSPCs and neurons (46.5% ßIII-tubulin+ cells). Gamma-aminobutyric acid (GABA)ergic and dopaminergic/noradrenergic neurons were also observed, along with a network of synaptic proteins. The ES-NSPCs expressed matriptase and secreted MMP-2/9, with MMP-2 activity increasing along time. Fibronectin, laminin and collagen type IV deposition was also detected. Fibrin gels prepared with higher fibrinogen concentrations (8/10 mg/ml) were less permissive to neurite extension and neuronal differentiation, possibly owing to their smaller pore area and higher rigidity. Overall, it is shown that ES-NSPCs within fibrin are able to establish neuronal networks and to remodel fibrin through MMP secretion and extracellular matrix (ECM) deposition. This three-dimensional (3D) culture system was also shown to support cell viability, neuronal differentiation and ECM deposition of human ES-NSPCs. The settled 3D platform is expected to constitute a valuable tool to develop fibrin-based hydrogels for ES-NSPC delivery into the injured central nervous system. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Extracellular Matrix/metabolism , Fibrin/pharmacology , Hydrogels/pharmacology , Nerve Net/physiology , Neural Stem Cells/cytology , Animals , Cell Count , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Extracellular Matrix/drug effects , Gels , Humans , Matrix Metalloproteinases/metabolism , Mice , Nerve Net/drug effects , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Phenotype , Serine Endopeptidases/metabolism
8.
Biomaterials ; 53: 566-73, 2015.
Article in English | MEDLINE | ID: mdl-25890752

ABSTRACT

In our search for immunomodulatory biomaterials capable of modulating the inflammatory response through M2 macrophage polarization, we report here on a new strategy that resulted from the incorporation of resolvin D1 (RvD1), a pro-resolution lipid mediator in porous 3D chitosan (Ch) scaffolds, followed by its lyophilisation. We have investigated the inflammatory response caused by this biomaterial in vivo using a mouse air-pouch model of inflammation. We found that this developed material caused a decrease in inflammatory cells recruited to the implant site, together with higher numbers of F4/80(+)/CD206(+) cells (M2 macrophages) and lower numbers of F4/80(+)/CCR7(+) cells (M1 macrophages). It also induced a general decrease in pro-inflammatory cytokines, and caused a decrease in the inflammatory cells observed around and within the implanted scaffolds, when compared with Ch alone or Ch not submitted to lyophilisation after RvD1 incorporation. Our results demonstrate that we were able to develop an immunomodulating biomaterial that triggers a shift in the macrophage response towards a M2 reparative response that will be advantageous for the host.


Subject(s)
Adjuvants, Immunologic/pharmacology , Biocompatible Materials , Docosahexaenoic Acids/pharmacology , Inflammation/therapy , Animals , Cytokines/biosynthesis , Mice , Tissue Scaffolds
9.
Biomaterials ; 37: 116-23, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25453942

ABSTRACT

Tissue engineering and regenerative medicine have created a demand for biomaterials with specific functions such as the ability to modify the host immune response. The objective of this study was to evaluate the effect of two different pro-resolution lipid mediators, lipoxin A4 (LxA4) and resolvin D1 (RvD1), in the modulation of the inflammatory response to biomaterials through M2 macrophage polarization. This was investigated in vivo using a mouse air-pouch model of inflammation. Our results demonstrated that both LxA4 and RvD1 are able to shift the macrophage response to implanted Ch scaffolds to an M2 reparative response. The injection of these pro-resolution mediators caused a decrease in inflammatory cells recruited to the implant site together with higher numbers of F4/80(+)/CD206(+) cells (M2 macrophages) and lower numbers of F4/80(+)/CCR7(+) cells (M1 macrophages); it also induced a general decrease in several pro-inflammatory cytokines; and caused a significant decrease in the thickness and area of the fibrous capsule formed around the implanted scaffolds. In conclusion, the use of either LxA4 or RvD1 allowed the in vivo control of macrophage phenotypic profile and thus may play a significant role in regenerative medicine applications, namely through modulation of the inflammatory response.


Subject(s)
Cell Polarity/drug effects , Chitosan/pharmacology , Docosahexaenoic Acids/pharmacology , Inflammation/pathology , Lipoxins/pharmacology , Macrophages/pathology , Animals , Biomarkers/metabolism , Cytokines/biosynthesis , Decapodiformes , Flow Cytometry , Implants, Experimental , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred BALB C
10.
Biomaterials ; 34(38): 9952-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24074837

ABSTRACT

Macrophages are a key cell in the host response to implants and can be polarized into different phenotypes capable of inducing both detrimental and beneficial outcomes in tissue repair and remodeling, being important in tissue engineering and regenerative medicine. The objective of this study was to evaluate the macrophage response to 3D porous chitosan (Ch) scaffolds with different degrees of acetylation (DA, 5% and 15%). The M1/M2 phenotypic polarization profile of macrophages was investigated in vivo using a rodent air-pouch model. Our results show that the DA affects the macrophage response. Ch scaffolds with DA 5% induced the adhesion of lower numbers of inflammatory cells, being the M2 the predominant phenotypic profile among the adherent macrophages. In the inflammatory exudates F4/80(+)/CD206(+) cells (M2 macrophages) appeared in higher numbers then F4/80(+)/CCR7(+) cells (M1 macrophages), in addition, lower levels of pro-inflammatory cytokines together with higher levels of anti-inflammatory cytokines were found. Ch scaffolds with DA 15% showed opposite results, since M1 were the predominant macrophages both adherent to the scaffold and in the exudates, together with high levels of pro-inflammatory cytokines. In conclusion, Ch scaffolds with DA 5% induced a benign M2 anti-inflammatory macrophage response, whereas Ch scaffolds with DA 15% caused a macrophage M1 pro-inflammatory response.


Subject(s)
Chitosan/chemistry , Macrophages/cytology , Macrophages/metabolism , Tissue Scaffolds/chemistry , Animals , Flow Cytometry , Inflammation/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Tissue Engineering/methods , Tumor Necrosis Factor-alpha/metabolism
11.
Int J Pharm ; 454(1): 116-24, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23850814

ABSTRACT

Chitosan microspheres have been explored for pharmaceutical applications, namely as a drug delivery systems for Helicobacter pylori gastric infection treatment, due to their mucoadhesive capacity. In this study, a different application of chitosan microspheres is proposed aiming the creation of an H. pylori-binding system where, after oral administration, microspheres will capture and remove these bacteria from infected patients, taking advantage of their muco/bacterial adhesive process. However, mucoadhesion is influenced by the degree of crosslinking necessary to avoid microspheres dissolution in the acidic gastric environment. During this work, the effect of genipin crosslinking on the stability, size, charge and mucoadhesive properties of chitosan microspheres under acidic pH was studied. Chitosan microspheres with ∼170 µm were produced by ionotropic gelation and subsequently covalently crosslinked with genipin in different degrees. The crosslinking reaction was followed by infrared spectroscopy and time-lapse fluorescence microscopy, since we have demonstrated that the fluorescence intensity of chitosan microspheres increases with genipin chemical bonding to chitosan. Results showed that both the zeta potential and the swelling capacity of chitosan microspheres decrease with increasing crosslinking. When immersed in simulated gastric fluid (SGF) with pepsin for 7 days, chitosan microspheres crosslinked with 10mM of genipin for 1h did not dissolve and doubled their size to approximately 345 µm. Furthermore, they maintained their in vitro mucoadhesion to soluble gastric mucins at both pH tested (3.6 and 6.5) and presented an in vivo retention time of around 2h in the stomach of C57BL/6 mice.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Chitosan/administration & dosage , Gastric Mucins/chemistry , Helicobacter pylori/metabolism , Stomach/microbiology , Adhesiveness , Administration, Oral , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Chemistry, Pharmaceutical , Chitosan/chemistry , Chitosan/metabolism , Cross-Linking Reagents/chemistry , Drug Stability , Gastric Juice/chemistry , Gastric Mucins/metabolism , Hydrogen-Ion Concentration , Iridoids/chemistry , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Microspheres , Particle Size , Pepsin A/chemistry , Solubility , Spectroscopy, Fourier Transform Infrared , Technology, Pharmaceutical/methods , Time Factors , Time-Lapse Imaging
12.
Biomatter ; 3(2)2013.
Article in English | MEDLINE | ID: mdl-23635535

ABSTRACT

In this study, (125)I-radiolabelling was explored to follow the kinetics and isotherm of fibronectin (FN) adsorption to porous polymeric scaffolds, as well as to assess the elution and exchangeability of pre-adsorbed FN following incubation in serum-containing culture medium. Chitosan (CH) porous scaffolds with two different degrees of acetylation (DA 4% and 15%) were incubated in FN solutions with concentrations ranging from 5 to 50 µg/mL. The kinetic and isotherm of FN adsorption to CH were successfully followed using (125)I-FN as a tracer molecule. While on DA 4% the levels of adsorbed FN increased linearly with FN solution concentration, on DA 15% a saturation plateau was attained, and FN adsorbed amounts were significantly lower. These findings were supported by immunofluorescent studies that revealed, for the same FN solution concentration, higher levels of exposed cell-binding domains on DA 4% as compared with DA 15%. Following incubation in serum containing medium, DA 4% also revealed higher ability to exchange pre-adsorbed FN by new FN molecules from serum than DA 15%. In accordance, when assessing the efficacy of passively adsorbed FN to promote endothelial cell (EC) adhesion to CH, ECs were found to adhere at higher levels to DA 4% as compared with DA 15%, 5 µg/mL of FN being already efficient in promoting cell adhesion and cytoskeletal organization on CH with DA 4%. Taken together the results show that protein radiolabelling can be used as an effective tool to study protein adsorption to porous polymeric scaffolds, both from single and complex protein solutions.


Subject(s)
Biocompatible Materials/chemistry , Chitosan/chemistry , Fibronectins/pharmacokinetics , Isotope Labeling/methods , Adsorption , Cell Line , Endothelial Cells , Fibronectins/chemistry , Humans , Iodine Radioisotopes , Kinetics , Porosity , Surface Properties , Tissue Scaffolds
13.
J Biomed Mater Res A ; 93(1): 20-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19484769

ABSTRACT

The effect of the degree of acetylation (DA) of 3D chitosan (Ch) scaffolds on the inflammatory reaction was investigated. Chitosan porous scaffolds with DAs of 4 and 15% were implanted using a subcutaneous air-pouch model of inflammation. The initial acute inflammatory response was evaluated 24 and 48 h after implantation. To characterize the initial response, the recruitment and adhesion of inflammatory cells to the implant site was studied. The fibrous capsule formation and the infiltration of inflammatory cells within the scaffolds were evaluated for longer implantation times (2 and 4 weeks). Chitosan with DA 15% attracted the highest number of leukocytes to the implant site. High numbers of adherent inflammatory cells were also observed in this material. For longer implantation periods Ch scaffolds with a DA of 15% induced the formation of a thick fibrous capsule and a high infiltration of inflammatory cells within the scaffold. Our results indicate that the biological response to implanted Ch scaffolds was influenced by the DA. Chitosan with a DA of 15% induce a more intense inflammatory response when compared with DA 4% Ch. Because inflammation and healing are interrelated, this result may provide clues for the relative importance of acetyl and amine functional groups in tissue repair and regeneration.


Subject(s)
Chitosan/adverse effects , Inflammation/chemically induced , Inflammation/pathology , Tissue Scaffolds/adverse effects , Tissue Scaffolds/chemistry , Acetylation/drug effects , Animals , Cell Adhesion/drug effects , Chitosan/immunology , Freeze Drying , Implants, Experimental , Leukocyte Count , Male , Mice , Mice, Inbred BALB C , Microscopy, Electron, Scanning , Porosity , Prosthesis Implantation , Subcutaneous Tissue/drug effects , Subcutaneous Tissue/pathology , Surface Properties/drug effects
14.
Biomaterials ; 30(29): 5465-75, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19615736

ABSTRACT

Chitosan (Ch) porous matrices were investigated regarding their ability to be colonized by human microvascular endothelial cells (HPMEC-ST1.6R cell line) and macrovascular endothelial cells namely HUVECs. Specifically we assessed if previous incubation of Ch in a fibronectin (FN) solution was effective in promoting endothelial cell (EC) adhesion to Ch matrices with different degrees of acetylation (DAs). Upon FN physiadsorption, marked differences were found between the two DAs investigated, namely DA 4% and 15%. While cell adhesion was impaired on Ch with DA 15%, ECs were able to not only adhere to Ch with DA 4%, but also to spread and colonize the scaffolds, with retention of the EC phenotype and angiogenic potential. To explain the observed differences between the two DAs, protein adsorption studies using (125)I-FN and immunofluorescent labelling of FN cell-binding domains were carried out. In agreement with the higher cell numbers found, scaffolds with DA 4% revealed a higher number of exposed FN cell-binding domains as well as greater ability to adsorb FN and to retain and exchange adsorbed FN in the presence of competitive proteins. These findings suggest that the DA is a key parameter modulating EC adhesion to FN-coated Ch by influencing the adsorbed protein layer.


Subject(s)
Chitosan/chemistry , Endothelial Cells/cytology , Endothelial Cells/physiology , Fibronectins/chemistry , Fibronectins/pharmacology , Neovascularization, Physiologic/physiology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Line , Cell Proliferation , Cell Size , Cell Survival , Endothelial Cells/drug effects , Humans , Materials Testing , Porosity
15.
Tissue Eng Part A ; 15(3): 635-43, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18759673

ABSTRACT

The use of biomaterials to replace lost bone has been a common practice for decades. More recently, the demands for bone repair and regeneration have pushed research into the use of cultured cells and growth factors in association with these materials. Here we report a novel approach to engineer new bone using a transient cartilage scaffold to induce endochondral ossification. Chondrocyte/chitosan scaffolds (both a transient cartilage scaffold-experimental-and a permanent cartilage scaffold-control) were prepared and implanted subcutaneously in nude mice. Bone formation was evaluated over a period of 5 months. Mineralization was assessed by Faxitron, micro computed tomography, backscatter electrons, and Fourier transform infrared spectroscopy analyses. Histological analysis provided further information on tissue changes in and around the implanted scaffolds. The deposition of ectopic bone was detected in the surface of the experimental implants as early as 1 month after implantation. After 3 months, bone trabeculae and bone marrow cavities were formed inside the scaffolds. The bone deposited was similar to the bone of the mice vertebra. Interestingly, no bone formation was observed in control implants. In conclusion, an engineered transient cartilage template carries all the signals necessary to induce endochondral bone formation in vivo.


Subject(s)
Bone and Bones/physiology , Cartilage/physiology , Tissue Engineering/methods , Animals , Bone and Bones/drug effects , Calcification, Physiologic/drug effects , Cartilage/drug effects , Chick Embryo , Chitosan/pharmacology , Chondrocytes/cytology , Chondrocytes/drug effects , Male , Mice , Mice, Nude , Microscopy, Electron, Scanning , Minerals/metabolism , Prosthesis Implantation , Spectroscopy, Fourier Transform Infrared , Tissue Scaffolds
16.
Tissue Eng Part A ; 15(3): 625-34, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18759672

ABSTRACT

Chitosan scaffolds have been shown to possess biological and mechanical properties suitable for tissue engineering and clinical applications. In the present work, chitosan sponges were evaluated regarding their ability to support cartilage cell proliferation and maturation, which are the first steps in endochondral bone formation. Chitosan sponges were seeded with chondrocytes isolated from chicken embryo sterna. Chondrocyte/chitosan constructs were cultured for 20 days, and treated with retinoic acid (RA) to induce chondrocyte maturation and matrix synthesis. At different time points, samples were collected for microscopic, histological, biochemical, and mechanical analyses. Results show chondrocyte attachment, proliferation, and abundant matrix synthesis, completely obliterating the pores of the sponges. RA treatment caused chondrocyte hypertrophy, characterized by the presence of type X collagen in the extracellular matrix and increased alkaline phosphatase activity. In addition, hypertrophy markedly changed the mechanical properties of the chondrocyte/chitosan constructs. In conclusion, we have developed chitosan sponges with adequate pore structure and mechanical properties to serve as a support for hypertrophic chondrocytes. In parallel studies, we have evaluated the ability of this mature cartilage scaffold to induce endochondral ossification.


Subject(s)
Bone and Bones/physiology , Cartilage/physiology , Tissue Engineering/methods , Alkaline Phosphatase/metabolism , Animals , Biomechanical Phenomena , Bone and Bones/drug effects , Cartilage/cytology , Cartilage/drug effects , Cells, Cultured , Chick Embryo , Chitosan/pharmacology , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/enzymology , Collagen Type X/metabolism , DNA/metabolism , Gene Expression Profiling , Porifera , Porosity/drug effects , Tissue Scaffolds , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...