Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Biomed Mater ; 18(6)2023 09 11.
Article in English | MEDLINE | ID: mdl-37604159

ABSTRACT

BACKGROUND: volumetric muscle loss (VML) is a traumatic massive loss of muscular tissue which frequently leads to amputation, limb loss, or lifetime disability. The current medical intervention is limited to autologous tissue transfer, which usually leads to non-functional tissue recovery. Tissue engineering holds a huge promise for functional recovery. METHODS: in this work, we evaluated the potential of human adipose-derived mesenchymal stem cells (hASCs) pre-cultured in gellan gum based spongy-like hydrogels (SLHs). RESULTS: in vitro, hASCs were spreading, proliferating, and releasing growth factors and cytokines (i.e. fibroblast growth factor, hepatocyte growth factor, insulin-like growth factor 1, interleukin-6 (IL-6), IL-8, IL-10, vascular endothelial growth factor) important for muscular regeneration. After implantation into a volumetric muscle loss (VML) mouse model, implants were degrading overtime, entirely integrating into the host between 4 and 8 weeks. In both SLH and SLH + hASCs defects, infiltrated cells were observed inside constructs associated with matrix deposition. Also, minimal collagen deposition was marginally observed around the constructs along both time-points. Neovascularization (CD31+vessels) and neoinnervation (ß-III tubulin+bundles) were significantly detected in the SLH + hASCs group, in relation to the SHAM (empty lesion). A higher density ofα-SA+and MYH7+cells were found in the injury site among all different experimental groups, at both time-points, in relation to the SHAM. The levels ofα-SA, MyoD1, and myosin heavy chain proteins were moderately increased in the SLH + hASCs group after 4 weeks, and in the hASCs group after 8 weeks, in relation to the SHAM. CONCLUSIONS: taken together, defects treated with hASCs-laden SLH promoted angiogenesis, neoinnervation, and the expression of myogenic proteins.


Subject(s)
Polysaccharides, Bacterial , Vascular Endothelial Growth Factor A , Animals , Mice , Humans , Cytokines , Muscles
2.
Int J Biol Macromol ; 249: 125957, 2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37499705

ABSTRACT

Heparin is the most common anticoagulant used in clinical practice but shows some downsides such as short half-life (for the high molecular weight heparin) and secondary effects. On the other hand, its low molecular weight analogue cannot be neutralized with protamine, and therefore cannot be used in some treatments. To address these issues, we conjugated polyethylene glycol (PEG) to heparin reducing end (end-on) via oxime ligation and studied the interactions of the conjugate (Hep-b-PEG) with antithrombin III (AT) and protamine. Isothermal titration calorimetry showed that Hep-b-PEG maintains the affinity to AT. Dynamic light scattering demonstrated that the Hep-b-PEG formed colloidal stable nanocomplexes with protamine instead of large multi-molecular aggregates, associated with heparin side effects. The in vitro (human plasma) and in vivo experiments (Sprague Dawley rats) evidenced an extended half-life and higher anticoagulant activity of the conjugate when compared to unmodified heparin.


Subject(s)
Heparin , Protamines , Animals , Rats , Humans , Heparin/adverse effects , Protamines/chemistry , Rats, Sprague-Dawley , Anticoagulants/pharmacology , Anticoagulants/chemistry
3.
Acta Biomater ; 143: 282-294, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35278687

ABSTRACT

Moderate muscular injuries that exceed muscular tissue's auto-healing capacity are still a topic of noteworthy concern. Tissue engineering appeared as a promising therapeutic strategy capable of overcoming this unmet clinical need. To attain such goal, herein we propose an in situ-crosslinking gellan gum (GG)-based hydrogel tethered with a skeletal muscle-inspired laminin-derived peptide RKRLQVQLSIRTC(Q) and encapsulated with skeletal muscle cells (SMCs). Pre-hydrogel solutions presented decreasing shear viscosity with increasing shear rate and shear stress, and required low forces for extrusion, validating their injectability. The GGDVS hydrogel was functionalized with Q-peptide with 30% of efficiency. C2C12 were able to adhere to the developed hydrogel, remained living and spreading 7 days post-encapsulation. Q-peptide release studies indicated that 25% of the unbound peptide can be released from the hydrogels up to 7 days, dependent on the hydrogel formulation. Treatment of a chemically-induced muscular lesion in mice with an injection of C2C12-laden hydrogels improved myogenesis, primarily promoted by the C2C12. In accordance, a high density of myoblasts (α-SA+ and MYH7+) were localized in tissues treated with the C2C12 (alone or encapsulated in the hydrogel). α-SA protein levels were significantly increased 8 weeks post-treatment with C2C12-laden hydrogels and MHC protein levels were increased in all experimental groups 4 weeks post-treatment, in relation to the SHAM. Neovascularization and neoinnervation was also detected in the defects. Altogether, this study indicates that C2C12-laden hydrogels hold great potential for skeletal muscle regeneration. STATEMENT OF SIGNIFICANCE: We developed an injectable gellan gum-based hydrogel for delivering C2C12 into localized myopathic model. The gellan gum was biofunctinalized with laminin-derived peptide to mimic the native muscular ECM. In addition, hydrogel was physically tuned to mimic the mechanical properties of native tissue. To the best of our knowledge, this formula was used for the first time under the context of skeletal muscle tissue regeneration. The injectability of the developed hydrogel provided non-invasive administration method, combined with a reliable microenvironment that can host C2C12 with nominal inflammation, indicated by the survival and adhesion of encapsulated cells post-injection. The treatment of skeletal muscle defect with the cell-laden hydrogel approach significantly enhanced the regeneration of localized muscular trauma.


Subject(s)
Hydrogels , Laminin , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Laminin/pharmacology , Mice , Muscle, Skeletal , Myoblasts , Peptides , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/pharmacology , Tissue Engineering/methods
4.
ACS Biomater Sci Eng ; 7(10): 4898-4913, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34533303

ABSTRACT

Cell encapsulation strategies using hydrogel beads have been considered as an alternative to immunosuppression in cell-based therapies. They rely on layer-by-layer (LbL) deposition of polymers to tune beads' permeability, creating a physical barrier to the host immune system. However, the LbL approach can also create diffusion barriers, hampering the flow of essential nutrients and therapeutic cell products. In this work, the polyelectrolyte complex (PEC) methodology was used to circumvent the drawbacks of the LbL strategy by inducing hydrogel bead formation through the interaction of anionic methacrylated gellan gum (GG-MA) with cationic poly-l-lysine (PLL). The interfacial complexation between both polymers resulted in beads with a cell-friendly GG-MA hydrogel core surrounded by a PEC semipermeable membrane. The beads showed great in vitro stability over time, a semi-permeable behavior, and supported human adipose-derived stem cell encapsulation. Additionally, and regarding immune recognition, the in vitro and in vivo studies pointed out that the hydrogel beads behave as an immunocompatible system. Overall, the engineered beads showed great potential for hydrogel-mediated cell therapies, when immunoprotection is required, as when treating different metabolic disorders.


Subject(s)
Polylysine , Polysaccharides, Bacterial , Humans , Hydrogels , Polyelectrolytes
5.
Mater Sci Eng C Mater Biol Appl ; 121: 111845, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33579481

ABSTRACT

Rheumatoid arthritis is a chronic autoimmune disease characterised by joint synovial inflammation, along with cartilage and bone tissue destruction. Dendrimers can offer new opportunities as drug delivery systems of molecules of interest. Herein we aimed to develop poly(amidoamine) dendrimers (PAMAM), functionalised with chondroitin sulphate (CS), lined with anti-TNF α antibodies (Abs) to provide anti-inflammatory properties. Physicochemical characterisation demonstrated that anti-TNFα Abs-CS/PAMAM dendrimer NPs were successfully produced. The in vitro studies revealed that CS/PAMAM dendrimer NPs did not affect the ATDC5 and THP-1 cell lines' metabolic activity and proliferation, presenting good cytocompatibility and hemocompatibility. Moreover, anti-TNFα Abs-CS/PAMAM dendrimer NPs showed suitable TNF α capture capacity, making them appealing for new immunotherapies in RA patients.


Subject(s)
Arthritis, Rheumatoid , Dendrimers , Arthritis, Rheumatoid/drug therapy , Chondroitin Sulfates , Humans , Tumor Necrosis Factor-alpha
6.
Macromol Biosci ; 21(4): e2000425, 2021 04.
Article in English | MEDLINE | ID: mdl-33522095

ABSTRACT

Hydrogels, being capable of mimicking the extracellular matrix composition of tissues, are greatly used as artificial matrices in tissue engineering applications. In this study, the generation of horseradish peroxidase (HRP)-crosslinked silk fibroin (SF) hydrogels, using calcium peroxide as oxidizer is reported. The proposed fast forming calcium-containing SF hydrogels spontaneously undergo SF conformational changes from random coil to ß-sheet during time, exhibiting ionic, and pH stimuli responsiveness. In vitro response shows calcium-containing SF hydrogels' encapsulation properties and their ability to promote SaOs-2 tumor cells death after 10 days of culturing, upon complete ß-sheet conformation transition. Calcium-containing SF hydrogels' angiogenic potential investigated in an in ovo chick chorioallantoic membrane (CAM) assay, show a high number of converging blood vessels as compared to the negative control, although no endothelial cells infiltration is observed. The in vivo response evaluated in subcutaneous implantation in CD1 and nude NCD1 mice shows that calcium-containing SF hydrogels are stable up to 6 weeks after implantation. However, an increased number of dead cells are also present in the surrounding tissue. The results suggest the potential of calcium-containing SF hydrogels to be used as novel in situ therapeutics for bone cancer treatment applications, particularly to osteosarcoma.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Fibroins/chemistry , Horseradish Peroxidase/chemistry , Hydrogels , Animals , Bone and Bones/metabolism , Calcium , Cell Line, Tumor , Chorioallantoic Membrane/metabolism , Humans , Hydrogels/chemistry , Hydrogen-Ion Concentration , Mice , Neovascularization, Pathologic , Protein Conformation , Silk/metabolism , Tissue Engineering
7.
Adv Exp Med Biol ; 1230: 65-86, 2020.
Article in English | MEDLINE | ID: mdl-32285365

ABSTRACT

Over the past years, important progresses have been made in the field of tissue engineering. Many of the early trials to improve the development of an engineered tissue construct were centered on the concept of seeding cells onto biomaterial scaffold. By means of innovative manufacturing machineries, the conception of a preformed scaffold became possible. Nowadays, several tissue engineering challenges are associated with applying this scaffold technology to one vital organ construct: liver. The development of microscale tissue ("micro-tissue") constructs to mimic partially the complex structure-function interactions of liver parenchyma have been obtained through the engineering of sophisticated biomaterial scaffolds, liver-cell sources, and in vitro culture techniques. For in vitro applications, micro-tissue constructs are being upgraded into cell-based assays for testing acute, chronic and idiosyncratic toxicities of drugs or pathogens. The present chapter will focus on the biomaterials currently used for the development of in vitro liver constructs as well as the description of the microfluidic-based models that show great promise for liver regenerative medicine approaches.


Subject(s)
Biocompatible Materials , Liver , Microfluidics , Models, Biological , Humans , Regenerative Medicine , Tissue Engineering , Tissue Scaffolds
8.
Biofabrication ; 12(2): 025028, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32069441

ABSTRACT

The meniscus has critical functions in the knee joint kinematics and homeostasis. Injuries of the meniscus are frequent, and the lack of a functional meniscus between the femur and tibial plateau can cause articular cartilage degeneration leading to osteoarthritis development and progression. Regeneration of meniscus tissue has outstanding challenges to be addressed. In the current study, novel Entrapped in cage (EiC) scaffolds of 3D-printed polycaprolactone (PCL) and porous silk fibroin were proposed for meniscus tissue engineering. As confirmed by micro-structural analysis the entrapment of silk fibroin was successful, and all scaffolds had excellent interconnectivity (≥99%). The EiC scaffolds had more favorable micro-structure compared with the PCL cage scaffolds by improving the pore size while keeping the interconnectivity almost the same. When compared with the PCL cage, the entrapment of porous silk fibroin into the PCL cage decreased the high compressive modulus in a favorable matter in the wet state thanks to the silk fibroin's high swelling properties. The in vitro studies with human stem cells or meniscocytes seeded constructs, demonstrated that the EiC scaffolds had superior cell adhesion, metabolic activity, and proliferation compared to the PCL cage scaffolds. Upon subcutaneous implantation of scaffolds in nude mice, all groups were free of adverse incidents, and mildly invaded by inflammatory cells with neovascularization, while the EiC scaffolds showed better tissue infiltration. The results of this work indicated that the EiC scaffolds of PCL and silk fibroin are favorable for meniscus tissue engineering, and the findings are encouraging for further studies using a larger animal model.


Subject(s)
Fibroins/chemistry , Polyesters/chemistry , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Adhesion/drug effects , Cell Survival/drug effects , Humans , Male , Meniscus/cytology , Meniscus/metabolism , Meniscus/transplantation , Mice , Mice, Nude , Porosity , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/metabolism
9.
Adv Healthc Mater ; 9(5): e1901435, 2020 03.
Article in English | MEDLINE | ID: mdl-31977159

ABSTRACT

Liver diseases contribute markedly to the global burden of mortality and disease. The limited organ disposal for orthotopic liver transplantation results in a continuing need for alternative strategies. Over the past years, important progress has been made in the field of tissue engineering (TE). Many of the early trials to improve the development of an engineered tissue construct are based on seeding cells onto biomaterial scaffolds. Nowadays, several TE approaches have been developed and are applied to one vital organ: the liver. Essential elements must be considered in liver TE-cells and culturing systems, bioactive agents or growth factors (GF), and biomaterials and processing methods. The potential of hepatocytes, mesenchymal stem cells, and others as cell sources is demonstrated. They need engineered biomaterial-based scaffolds with perfect biocompatibility and bioactivity to support cell proliferation and hepatic differentiation as well as allowing extracellular matrix deposition and vascularization. Moreover, they require a microenvironment provided using conventional or advanced processing technologies in order to supply oxygen, nutrients, and GF. Herein the biomaterials and the conventional and advanced processing technologies, including cell-sheets process, 3D bioprinting, and microfluidic systems, as well as the future trends in these major fields are discussed.


Subject(s)
Biocompatible Materials , Bioprinting , Extracellular Matrix , Liver Regeneration , Tissue Engineering , Tissue Scaffolds
10.
Acta Biomater ; 93: 74-85, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30708066

ABSTRACT

In this study, methacrylated gellan-gum (GG-MA) heteropolysaccharide is proposed as a hydrogel for drug delivery and bone tissue engineering applications. Calcium-enriched beads obtained from the crosslinking of 1% (w/v) GG-MA solutions with 0.1 MCaCl2 were investigated, considering their intrinsic capacity to promote self-mineralization by ion binding and deposition. Indeed, when immersed in a physiological environment, the Ca-enriched beads promoted the development of a bone-like apatite layer, as confirmed by EDS and XRD chemical analysis. Additionally, the mild production process is compatible with drugs incorporation and release. After encapsulation, Dextran with different molecular weights as well as Dexamethasone 21-phosphate were efficiently released to the surrounding environment. The engineered system was also evaluated considering its biocompatibility, by means of qualitative determination of total complement activation, macrophage proliferation, cytokine release and in vitro cell culture. These experiments showed that the developed hydrogels may not stimulate a disproportionate pro-inflammatory reaction once transplanted. At last, when implanted subcutaneously in CD1 male mice up to 8 weeks, the beads were completely calcified, and no inflammatory reaction was observed. Summing up, these results show that calcium-enriched GG-MA hydrogel beads hold great potential as news tools for bone tissue regeneration and local drug delivery applications. STATEMENT OF SIGNIFICANCE: This work describes a low-cost and straightforward strategy to prepare bioactive methacrylated gellan gum (GG-MA) hydrogels, which can be used as drug delivery systems. GG-MA is a highly anionic polymer, that can be crosslinked with divalent ions, as calcium. Taking advantage of this feature, it was possible to prepare Ca-enriched GG-MA hydrogel beads. These beads display a bioactive behavior, since they promote apatite deposition when placed in physiological conditions. Studies on the immune response suggest that the developed beads do not trigger severe immune responses. Importantly, the mild processing method render these beads compliant with drug delivery strategies, paving the way for the application of dual-functional materials on bone tissue engineering.


Subject(s)
Calcification, Physiologic/drug effects , Calcium , Hydrogels , Materials Testing , Polysaccharides, Bacterial , Tissue Engineering , Adolescent , Adult , Animals , Bone and Bones/metabolism , Calcium/chemistry , Calcium/pharmacology , Female , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Male , Methacrylates/chemistry , Methacrylates/pharmacology , Mice , Middle Aged , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/pharmacology
11.
Adv Healthc Mater ; 7(17): e1800186, 2018 09.
Article in English | MEDLINE | ID: mdl-29999601

ABSTRACT

Hollow tubular conduits (TCs) with tunable architecture and biological properties are in great need for modulating cell functions and drug delivery in guided tissue regeneration. Here, a new methodology to produce enzymatically cross-linked silk fibroin TCs is described, which takes advantage of the tyrosine groups present in silk structure that are known to allow the formation of a covalently cross-linked hydrogel. Three different processing methods are used as a final step to modulate the properties of the silk-based TCs. This approach allows to virtually adjust any characteristic of the final TCs. The final microstructure ranges from a nonporous to a highly porous network, allowing the TCs to be selectively porous to 4 kDa molecules, but not to human skin fibroblasts. Mechanical properties are dependent both on the processing method and thickness of the TCs. Bioactivity is observed after 30 days of immersion in simulated body fluid only for the TCs submitted to a drying processing method (50 °C). The in vivo study performed in mice demonstrates the good biocompatibility of the TCs. The enzymatically cross-linked silk fibroin TCs are versatile and have adjustable characteristics that can be exploited in a variety of biomedical applications, particularly in guidance of peripheral nerve regeneration.


Subject(s)
Fibroins/chemistry , Guided Tissue Regeneration/methods , Silk/chemistry , Animals , Humans , Mice , Microscopy, Electron, Scanning
12.
Adv Exp Med Biol ; 1059: 423-439, 2018.
Article in English | MEDLINE | ID: mdl-29736585

ABSTRACT

Animal assays represent an important stage between in vitro studies and human clinical applications. These models are crucial for biomedical research and regenerative medicine studies, as these offer precious information for systematically assessing the efficacy and risks of recently created biomaterials, medical devices, drugs, and therapeutic modalities prior to initiation of human clinical trials. Therefore, selecting a suitable experimental model for tissue engineering purposes is essential to establish valid conclusions. However, it remains important to be conscious of the advantages and limitations of the various small and large animal models frequently used for biomedical research as well as the different challenges encountered in extrapolating data obtained from animal studies and the risks of misinterpretation. This chapter discusses the various small animal model strategies used for osteochondral defect repair. Particular emphasis will be placed on analyzing the materials and strategies used in each model.


Subject(s)
Bone Diseases/therapy , Cartilage Diseases/therapy , Guinea Pigs , Materials Testing/methods , Mice , Models, Animal , Rabbits , Rats , Tissue Engineering/methods , Animals , Biocompatible Materials/therapeutic use , Bone Diseases/surgery , Cartilage Diseases/surgery , Drug Evaluation, Preclinical , Humans , Implants, Experimental , Intercellular Signaling Peptides and Proteins/therapeutic use , Species Specificity , Stem Cell Transplantation , Tissue Scaffolds
13.
Adv Exp Med Biol ; 1058: 391-413, 2018.
Article in English | MEDLINE | ID: mdl-29691832

ABSTRACT

Osteochondral lesions are frequent and important causes of pain and disability. These lesions are induced by traumatic injuries or by diseases that affect both the cartilage surface and the subchondral bone. Due to the limited cartilage ability to regenerate and self-repair, these lesions tend to gradually worsen and progress towards osteoarthritis. The clinical, social, and economic impact of the osteochondral lesions is impressive and although therapeutic alternatives are under discussion, a consensus is not yet been achieved. Over the previous decade, new strategies based on innovative tissue engineering approaches have been developed with promising results. However, in order those products reach the market and help the actual patient in an effective manner, there is still a lot of work to be done. The current state of the implications, clinical aspects, and available treatments for this pathology, as well as the ongoing preclinical and clinical trials are presented in this chapter.


Subject(s)
Bone and Bones , Cartilage , Osteoarthritis , Tissue Engineering/methods , Animals , Bone and Bones/injuries , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Clinical Trials as Topic , Humans , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/therapy
14.
Acta Biomater ; 72: 167-181, 2018 05.
Article in English | MEDLINE | ID: mdl-29626700

ABSTRACT

Several processing technologies and engineering strategies have been combined to create scaffolds with superior performance for efficient tissue regeneration. Cartilage tissue is a good example of that, presenting limited self-healing capacity together with a high elasticity and load-bearing properties. In this work, novel porous silk fibroin (SF) scaffolds derived from horseradish peroxidase (HRP)-mediated crosslinking of highly concentrated aqueous SF solution (16 wt%) in combination with salt-leaching and freeze-drying methodologies were developed for articular cartilage tissue engineering (TE) applications. The HRP-crosslinked SF scaffolds presented high porosity (89.3 ±â€¯0.6%), wide pore distribution and high interconnectivity (95.9 ±â€¯0.8%). Moreover, a large swelling capacity and favorable degradation rate were observed up to 30 days, maintaining the porous-like structure and ß-sheet conformational integrity obtained with salt-leaching and freeze-drying processing. The in vitro studies supported human adipose-derived stem cells (hASCs) adhesion, proliferation, and high glycosaminoglycans (GAGs) synthesis under chondrogenic culture conditions. Furthermore, the chondrogenic differentiation of hASCs was assessed by the expression of chondrogenic-related markers (collagen type II, Sox-9 and Aggrecan) and deposition of cartilage-specific extracellular matrix for up to 28 days. The cartilage engineered constructs also presented structural integrity as their mechanical properties were improved after chondrogenic culturing. Subcutaneous implantation of the scaffolds in CD-1 mice demonstrated no necrosis or calcification, and deeply tissue ingrowth. Collectively, the structural properties and biological performance of these porous HRP-crosslinked SF scaffolds make them promising candidates for cartilage regeneration. STATEMENT OF SIGNIFICANCE: In cartilage tissue engineering (TE), several processing technologies have been combined to create scaffolds for efficient tissue repair. In our study, we propose novel silk fibroin (SF) scaffolds derived from enzymatically crosslinked SF hydrogels processed by salt-leaching and freeze-drying technologies, for articular cartilage applications. Though these scaffolds, we were able to combine the elastic properties of hydrogel-based systems, with the stability, resilience and controlled porosity of scaffolds processed via salt-leaching and freeze-drying technologies. SF protein has been extensively explored for TE applications, as a result of its mechanical strength, elasticity, biocompatibility, and biodegradability. Thus, the structural, mechanical and biological performance of the proposed scaffolds potentiates their use as three-dimensional matrices for cartilage regeneration.


Subject(s)
Cartilage/physiology , Chondrogenesis , Regeneration , Stem Cells/metabolism , Subcutaneous Fat/metabolism , Tissue Scaffolds/chemistry , Animals , Fibroins , Humans , Materials Testing , Mice , Mice, Inbred ICR , Stem Cells/cytology , Subcutaneous Fat/cytology , Tissue Engineering
15.
J Biomed Mater Res A ; 106(7): 1987-1996, 2018 07.
Article in English | MEDLINE | ID: mdl-29569326

ABSTRACT

Methacrylated gellan gum (GGMA) formulation is proposed as a second-generation hydrogel for controlled delivery of cartilage-forming cells into focal chondral lesions, allowing immediate in situ retention of cells and 3D filling of lesion volume, such approach deemed compatible with an arthroscopic procedure. Formulation optimization was carried out in vitro using chondrocytes and adipose mesenchymal stromal/stem cells (ASCs). A proof-of-concept in vivo study was conducted using a rabbit model with induced chondral lesions. Outcomes were compared with microfracture or non-treated control. Three grading scores were used to evaluate tissue repair after 8 weeks by macroscopic, histological and immunohistochemical analysis. Intense collagen type II and low collagen type I gene and protein expression were achieved in vitro by the ASC + GGMA formulation, in light with development of healthy chondral tissue. In vivo, this formulation promoted significantly superior de novo cartilage formation compared with the non-treated group. Maintenance of chondral height and integration with native tissue was further accomplished. The physicochemical properties of the proposed GGMA hydrogel exhibited highly favorable characteristics and biological performance both in vitro and in vivo, positioning itself as an attractive xeno-free biomaterial to be used with chondrogenic cells for a cost-effective treatment of focal chondral lesions. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 1987-1996, 2018.


Subject(s)
Cartilage, Articular/pathology , Hydrogels/pharmacology , Methacrylates/pharmacology , Polysaccharides, Bacterial/pharmacology , Wound Healing/drug effects , Adipose Tissue/cytology , Animals , Cartilage, Articular/drug effects , Chondrogenesis/drug effects , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Rabbits
16.
Biomaterials ; 123: 92-106, 2017 04.
Article in English | MEDLINE | ID: mdl-28161684

ABSTRACT

Bone loss in the craniofacial complex can been treated using several conventional therapeutic strategies that face many obstacles and limitations. In this work, novel three-dimensional (3D) biotextile architectures were developed as a possible strategy for flat bone regeneration applications. As a fully automated processing route, this strategy as potential to be easily industrialized. Silk fibroin (SF) yarns were processed into weft-knitted fabrics spaced by a monofilament of polyethylene terephthalate (PET). A comparative study with a similar 3D structure made entirely of PET was established. Highly porous scaffolds with homogeneous pore distribution were observed using micro-computed tomography analysis. The wet state dynamic mechanical analysis revealed a storage modulus In the frequency range tested, the storage modulus values obtained for SF-PET scaffolds were higher than for the PET scaffolds. Human adipose-derived stem cells (hASCs) cultured on the SF-PET spacer structures showed the typical pattern for ALP activity under osteogenic culture conditions. Osteogenic differentiation of hASCs on SF-PET and PET constructs was also observed by extracellular matrix mineralization and expression of osteogenic-related markers (osteocalcin, osteopontin and collagen type I) after 28 days of osteogenic culture, in comparison to the control basal medium. The quantification of convergent macroscopic blood vessels toward the scaffolds by a chick chorioallantoic membrane assay, showed higher angiogenic response induced by the SF-PET textile scaffolds than PET structures and gelatin sponge controls. Subcutaneous implantation in CD-1 mice revealed tissue ingrowth's accompanied by blood vessels infiltration in both spacer constructs. The structural adaptability of textile structures combined to the structural similarities of the 3D knitted spacer fabrics to craniofacial bone tissue and achieved biological performance, make these scaffolds a possible solution for tissue engineering approaches in this area.


Subject(s)
Bone Regeneration/physiology , Bone Substitutes/chemical synthesis , Mesenchymal Stem Cells/physiology , Osteoblasts/physiology , Osteogenesis/physiology , Silk/chemistry , Tissue Scaffolds , Anisotropy , Cell Differentiation/physiology , Cells, Cultured , Humans , Materials Testing , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Printing, Three-Dimensional , Textiles
17.
Biotechnol Bioeng ; 114(4): 717-739, 2017 04.
Article in English | MEDLINE | ID: mdl-27618194

ABSTRACT

Osteoarthritis (OA) affects a large number of the population, and its incidence is showing a growing trend with the increasing life span. OA is the most prevalent joint condition worldwide, and currently, there is no functional cure for it. This review seeks to briefly overview the management of knee OA concerning standardized pharmaceutical and clinical approaches, as well as the new biotechnological horizons of OA treatment. The potential of biomaterials and state of the art of advanced therapeutic approaches, such as cell and gene therapy focused primarily on cartilage regeneration are the main subjects of this review. Biotechnol. Bioeng. 2017;114: 717-739. © 2016 Wiley Periodicals, Inc.


Subject(s)
Osteoarthritis, Knee/therapy , Biocompatible Materials , Cartilage, Articular/cytology , Cartilage, Articular/physiology , Genetic Therapy , Humans , Tissue Engineering
18.
Sci Rep ; 6: 31037, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27485515

ABSTRACT

Protein-based hydrogels with distinct conformations which enable encapsulation or differentiation of cells are of great interest in 3D cancer research models. Conformational changes may cause macroscopic shifts in the hydrogels, allowing for its use as biosensors and drug carriers. In depth knowledge on how 3D conformational changes in proteins may affect cell fate and tumor formation is required. Thus, this study reports an enzymatically crosslinked silk fibroin (SF) hydrogel system that can undergo intrinsic conformation changes from random coil to ß-sheet conformation. In random coil status, the SF hydrogels are transparent, elastic, and present ionic strength and pH stimuli-responses. The random coil hydrogels become ß-sheet conformation after 10 days in vitro incubation and 14 days in vivo subcutaneous implantation in rat. When encapsulated with ATDC-5 cells, the random coil SF hydrogel promotes cell survival up to 7 days, whereas the subsequent ß-sheet transition induces cell apoptosis in vitro. HeLa cells are further incorporated in SF hydrogels and the constructs are investigated in vitro and in an in vivo chick chorioallantoic membrane model for tumor formation. In vivo, Angiogenesis and tumor formation are suppressed in SF hydrogels. Therefore, these hydrogels provide new insights for cancer research and uses of biomaterials.


Subject(s)
Biomimetic Materials , Fibroins , Hydrogels , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Chick Embryo , Fibroins/chemistry , Fibroins/pharmacology , HeLa Cells , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Mice , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Rats , Xenograft Model Antitumor Assays
19.
Clin Sci (Lond) ; 118(6): 411-20, 2009 Dec 14.
Article in English | MEDLINE | ID: mdl-19852755

ABSTRACT

Inhibition or blockade of HSCs (hepatic stellate cells), the main matrix-producing cells involved in the wound-healing response, represents an attractive strategy for the treatment of liver fibrosis. In vitro studies have shown that activation of AMPK (AMP-activated protein kinase), a key player in the regulation of cellular energy homoeostasis, inhibits proliferation of myofibroblasts derived from HSCs. If AMPK is a true regulator of fibrogenesis then defective AMPK activity would enhance fibrogenesis and hepatic fibrosis. To test this, in the present work, in vitro studies were performed on mouse primary HSCs treated or not with the AMPK activator AICAR (5-amino-4-imidazolecarboxamide ribonucleotide) or isolated from mice lacking the AMPKalpha1 catalytic subunit (AMPKalpha1-/-) or their littermates (AMPKalpha1+/+). Liver fibrosis was induced in vivo in AMPKalpha1-/- and AMPKalpha1+/+ mice by repeated injections of CCl(4) (carbon tetrachloride). During culture activation of HSCs, AMPK protein and activity significantly increased and regulatory AMPKgamma3 mRNA was specifically up-regulated. Stimulation of AMPK activity by AICAR inhibited HSC proliferation, as expected, as well as collagen alpha1(I) expression. Importantly, AMPKalpha1 deletion inhibited proliferation of HSCs, but not fibrogenesis, in vivo. Moreover, AMPKalpha1 deletion was not associated with enhanced CCl(4)-induced fibrosis in vivo. In conclusion, our present findings demonstrate that HSC transdifferentiation is associated with increased AMPK activity that could relate to the stabilization of AMPK complex by the gamma3 subunits. Activation of AMPK in HSCs inhibits in vitro fibrogenesis. By contrast, low AMPK activity does not prevent HSC activation in vitro nor in in vivo fibrosis.


Subject(s)
AMP-Activated Protein Kinases/deficiency , Liver Cirrhosis/enzymology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Animals , Cell Transdifferentiation , Cells, Cultured , Female , Hepatic Stellate Cells/cytology , Immunohistochemistry , Mice , RNA/metabolism , Ribonucleotides/metabolism
20.
J Hepatol ; 50(3): 489-500, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19155087

ABSTRACT

BACKGROUND/AIMS: Peroxisome proliferator-activated receptor gamma (PPARgamma) agonist drugs, like pioglitazone (PGZ), are proposed as treatments for steatohepatitis. Their mechanisms of action remain ill-clarified. METHODS: To test the hypothesis that PGZ improves steatohepatitis through adiponectin-dependent stimulation of AMPK and/or PPARalpha, mice lacking adiponectin (Adipo(-/-)) or the AMPKalpha1 catalytic subunit (AMPKalpha1(-/-)) or wild-type (Wt) mice were fed the methionine and choline deficient (MCD) diet, supplemented or not with PGZ. RESULTS: In Wt mice, PGZ increased circulating levels of adiponectin and reduced the severity of MCD-induced steatohepatitis but there was no evidence of activation of AMPK or PPARalpha and their downstream targets. By contrast, PGZ completely repressed nuclear translocation of SREBP-1c, a key transcription factor for de novo lipogenesis. This effect was lacking in Adipo(-/-) mice in which PGZ failed to prevent steatohepatitis. Surprisingly, AMPKalpha1(-/-) mice were resistant to MCD-induced steatohepatitis, a status also associated with repression of SREBP-1c. CONCLUSIONS: The preventive effect of PGZ on MCD-induced steatohepatitis depends on adiponectin upregulation but apparently does not involve AMPK or PPARalpha activation. The inhibition of SREBP-1c and dependent repression of lipogenesis are likely to participate in this effect. The mechanisms by which PGZ and adiponectin control SREBP-1c and inflammation remain to be elucidated.


Subject(s)
Adiponectin/physiology , Fatty Liver/prevention & control , Inflammation/prevention & control , Sterol Regulatory Element Binding Protein 1/antagonists & inhibitors , Thiazolidinediones/therapeutic use , AMP-Activated Protein Kinase Kinases , Adiponectin/deficiency , Animals , Choline Deficiency/complications , DNA Primers , Female , Liver/enzymology , Liver/pathology , Methionine/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Pioglitazone , Protein Kinases/deficiency , Protein Kinases/genetics , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...