Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Chem Rev ; 120(19): 10662-10694, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32302091

RESUMO

This review provides a detailed overview of the rapidly advancing field of biofabrication, particularly with regards to the use of photo-cross-linking (i.e., light-based) techniques. The major emphasis of this review is on the fundamentals of photo-cross-linking and key criteria identified for the successful design and implementation of photo-cross-linked bioinks and bioresins in extrusion-based and lithography-based bioprinting. The general mechanisms associated with photo-cross-linking (e.g., free-radical chain polymerization, thiol-ene, photomediated redox) of natural and synthetic materials are described to inform bioink and bioresin design, which includes the selection of polymers, functional group modifications, photoinitiators, and light sources that enable facile and cytocompatible photo-cross-linking. Depending on material selection and the bioprinting technique of interest, we describe the specific bioink or bioresin properties and criteria that must be achieved to ensure optimal printability and utility. Finally, examples of current state-of-the-art applications of light-based bioprinting for in vitro tissue models, tissue engineering, and regenerative medicine are provided to further motivate future opportunities within the bioprinting landscape that are facilitated with light.


Assuntos
Materiais Biocompatíveis/química , Bioimpressão , Reagentes de Ligações Cruzadas/química , Impressão Tridimensional , Engenharia Tecidual , Humanos , Processos Fotoquímicos
2.
J Nanobiotechnology ; 20(1): 141, 2022 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-35303876

RESUMO

With the gradual demographic shift toward an aging and obese society, an increasing number of patients are suffering from bone and cartilage injuries. However, conventional therapies are hindered by the defects of materials, failing to adequately stimulate the necessary cellular response to promote sufficient cartilage regeneration, bone remodeling and osseointegration. In recent years, the rapid development of nanomedicine has initiated a revolution in orthopedics, especially in tissue engineering and regenerative medicine, due to their capacity to effectively stimulate cellular responses on a nanoscale with enhanced drug loading efficiency, targeted capability, increased mechanical properties and improved uptake rate, resulting in an improved therapeutic effect. Therefore, a comprehensive review of advancements in nanomedicine for bone and cartilage diseases is timely and beneficial. This review firstly summarized the wide range of existing nanotechnology applications in the medical field. The progressive development of nano delivery systems in nanomedicine, including nanoparticles and biomimetic techniques, which are lacking in the current literature, is further described. More importantly, we also highlighted the research advancements of nanomedicine in bone and cartilage repair using the latest preclinical and clinical examples, and further discussed the research directions of nano-therapies in future clinical practice.


Assuntos
Osso e Ossos , Nanomedicina , Cartilagem , Humanos , Nanomedicina/métodos , Medicina Regenerativa/métodos , Engenharia Tecidual/métodos
3.
Adv Exp Med Biol ; 1078: 245-269, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30357627

RESUMO

Growth factors (GFs) are often a key component in tissue engineering and regenerative medicine approaches. In order to fully exploit the therapeutic potential of GFs, GF delivery vehicles have to meet a number of key design criteria such as providing localized delivery and mimicking the dynamic native GF expression levels and patterns. The use of biomaterials as delivery systems is the most successful strategy for controlled delivery and has been translated into different commercially available systems. However, the risk of side effects remains an issue, which is mainly attributed to insufficient control over the release profile. This book chapter reviews the current strategies, chemistries, materials and delivery vehicles employed to overcome the current limitations associated with GF therapies.


Assuntos
Sistemas de Liberação de Medicamentos , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Medicina Regenerativa , Engenharia Tecidual , Materiais Biocompatíveis , Humanos
4.
Eur Radiol ; 27(1): 384-392, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27165137

RESUMO

OBJECTIVES: To quantify iodine uptake in articular cartilage as a marker of glycosaminoglycan (GAG) content using multi-energy spectral CT. METHODS: We incubated a 25-mm strip of excised osteoarthritic human tibial plateau in 50 % ionic iodine contrast and imaged it using a small-animal spectral scanner with a cadmium telluride photon-processing detector to quantify the iodine through the thickness of the articular cartilage. We imaged both spectroscopic phantoms and osteoarthritic tibial plateau samples. The iodine distribution as an inverse marker of GAG content was presented in the form of 2D and 3D images after applying a basis material decomposition technique to separate iodine in cartilage from bone. We compared this result with a histological section stained for GAG. RESULTS: The iodine in cartilage could be distinguished from subchondral bone and quantified using multi-energy CT. The articular cartilage showed variation in iodine concentration throughout its thickness which appeared to be inversely related to GAG distribution observed in histological sections. CONCLUSIONS: Multi-energy CT can quantify ionic iodine contrast (as a marker of GAG content) within articular cartilage and distinguish it from bone by exploiting the energy-specific attenuation profiles of the associated materials. KEY POINTS: • Contrast-enhanced articular cartilage and subchondral bone can be distinguished using multi-energy CT. • Iodine as a marker of glycosaminoglycan content is quantifiable with multi-energy CT. • Multi-energy CT could track alterations in GAG content occurring in osteoarthritis.


Assuntos
Cartilagem Articular/diagnóstico por imagem , Glicosaminoglicanos/análise , Iodo/farmacocinética , Osteoartrite/diagnóstico por imagem , Tomografia Computadorizada por Raios X/métodos , Meios de Contraste/farmacocinética , Dissecação , Humanos , Osteoartrite/patologia , Imagens de Fantasmas , Tíbia/diagnóstico por imagem
5.
J Mater Sci Mater Med ; 25(1): 173-83, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24081382

RESUMO

The use of magnesium (Mg) as a biodegradable metallic replacement of permanent orthopaedic materials is a current topic of interest and investigation. The appropriate biocompatibility, elastic modulus and mechanical properties of Mg recommend its suitability for bone fracture fixation. However, the degradation rates of Mg can be rapid and unpredictable resulting in mass hydrogen production and potential loss of mechanical integrity. Thus the application of calcium phosphate coatings has been considered as a means of improving the degradation properties of Mg. Brushite and monetite are utilized and their degradation properties (alongside uncoated Mg controls) are assessed in an in vivo subcutaneous environment and the findings compared to their in vitro degradation behaviour in immersion tests. The current findings suggest monetite coatings have significant degradation protective effects compared to brushite coatings in vivo. Furthermore, it is postulated that an in vitro immersion test may be used as a tentative predictor of in vivo subcutaneous degradation behavior of calcium phosphate coated and uncoated Mg.


Assuntos
Implantes Absorvíveis , Fosfatos de Cálcio/química , Fosfatos de Cálcio/farmacocinética , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacocinética , Magnésio/química , Magnésio/farmacocinética , Animais , Corrosão , Masculino , Teste de Materiais , Microscopia Eletrônica de Varredura , Ratos , Ratos Endogâmicos Lew , Propriedades de Superfície
6.
Adv Biol (Weinh) ; 8(2): e2300448, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37953659

RESUMO

For effective translation of research from tissue engineering and regenerative medicine domains, the cell-instructive extracellular matrix (ECM) of specific tissues must be accurately realized. As adipose tissue is gaining traction as a biomaterial for soft tissue reconstruction, with highly variable clinical outcomes obtained, a quantitative investigation of the adipose tissue matrisome is overdue. In this study, the human adipose tissue matrisome is profiled using quantitative sequential windowed acquisition of all theoretical fragment ion spectra - mass spectrometry (SWATH-MS) proteomics across a cohort of 13 fat-grafting patients, to provide characterization of ECM proteins within the tissue, and to understand human population variation. There are considerable differences in the expression of matrisome proteins across the patient cohort, with age and lipoaspirate collection technique contributing to the greatest variation across the core matrisome. A high abundance of basement membrane proteins (collagen IV and heparan sulfate proteoglycan) is detected, as well as fibrillar collagens I and II, reflecting the hierarchical structure of the tissue. This study provides a comprehensive proteomic evaluation of the adipose tissue matrisome and contributes to an enhanced understanding of the influence of the matrisome in adipose-related pathologies by providing a healthy reference cohort and details an experimental pipeline that can be further exploited for future biomaterial development.


Assuntos
Matriz Extracelular , Proteômica , Humanos , Proteômica/métodos , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/análise , Proteínas da Matriz Extracelular/metabolismo , Materiais Biocompatíveis/análise , Materiais Biocompatíveis/metabolismo , Tecido Adiposo/química , Tecido Adiposo/metabolismo
7.
Biofabrication ; 16(2)2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38373325

RESUMO

The delivery of oxygen within tissue engineered constructs is essential for cell survivability; however, achieving this within larger biofabricated constructs poses a significant challenge. Efforts to overcome this limitation often involve the delivery of synthetic oxygen generating compounds. The application of some of these compounds is problematic for the biofabrication of living tissues due to inherent issues such as cytotoxicity, hyperoxia and limited structural stability due to oxygen inhibition of radical-based crosslinking processes. This study aims to develop an oxygen delivering system relying on natural-derived components which are cytocompatible, allow for photopolymerization and advanced biofabrication processes, and improve cell survivability under hypoxia (1% O2). We explore the binding of human hemoglobin (Hb) as a natural oxygen deposit within photopolymerizable allylated gelatin (GelAGE) hydrogels through the spontaneous complex formation of Hb with negatively charged biomolecules (heparin, hyaluronic acid, and bovine serum albumin). We systematically study the effect of biomolecule inclusion on cytotoxicity, hydrogel network properties, Hb incorporation efficiency, oxygen carrying capacity, cell viability, and compatibility with 3D-bioassembly processes within melt electrowritten (MEW) scaffolds. All biomolecules were successfully incorporated within GelAGE hydrogels, displaying controllable mechanical properties and cytocompatibility. Results demonstrated efficient and tailorable Hb incorporation within GelAGE-Heparin hydrogels. The developed system was compatible with microfluidics and photopolymerization processes, allowing for the production of GelAGE-Heparin-Hb spheres. Hb-loaded spheres were assembled into MEW polycaprolactone scaffolds, significantly increasing the local oxygen levels. Ultimately, cells within Hb-loaded constructs demonstrated good cell survivability under hypoxia. Taken together, we successfully developed a hydrogel system that retains Hb as a natural oxygen deposit post-photopolymerization, protecting Hb from free-radical oxidation while remaining compatible with biofabrication of large constructs. The developed GelAGE-Heparin-Hb system allows for physoxic oxygen delivery and thus possesses a vast potential for use across broad tissue engineering and biofabrication strategies to help eliminate cell death due to hypoxia.


Assuntos
Engenharia Tecidual , Alicerces Teciduais , Humanos , Alicerces Teciduais/química , Engenharia Tecidual/métodos , Hidrogéis/farmacologia , Hidrogéis/química , Hipóxia , Oxigênio , Heparina/farmacologia
8.
Biofabrication ; 16(4)2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39121873

RESUMO

Current biofabrication strategies are limited in their ability to replicate native shape-to-function relationships, that are dependent on adequate biomimicry of macroscale shape as well as size and microscale spatial heterogeneity, within cell-laden hydrogels. In this study, a novel diffusion-based microfluidics platform is presented that meets these needs in a two-step process. In the first step, a hydrogel-precursor solution is dispersed into a continuous oil phase within the microfluidics tubing. By adjusting the dispersed and oil phase flow rates, the physical architecture of hydrogel-precursor phases can be adjusted to generate spherical and plug-like structures, as well as continuous meter-long hydrogel-precursor phases (up to 1.75 m). The second step involves the controlled introduction a small molecule-containing aqueous phase through a T-shaped tube connector to enable controlled small molecule diffusion across the interface of the aqueous phase and hydrogel-precursor. Application of this system is demonstrated by diffusing co-initiator sodium persulfate (SPS) into hydrogel-precursor solutions, where the controlled SPS diffusion into the hydrogel-precursor and subsequent photo-polymerization allows for the formation of unique radial stiffness patterns across the shape- and size-controlled hydrogels, as well as allowing the formation of hollow hydrogels with controllable internal architectures. Mesenchymal stromal cells are successfully encapsulated within hollow hydrogels and hydrogels containing radial stiffness gradient and found to respond to the heterogeneity in stiffness through the yes-associated protein mechano-regulator. Finally, breast cancer cells are found to phenotypically switch in response to stiffness gradients, causing a shift in their ability to aggregate, which may have implications for metastasis. The diffusion-based microfluidics thus finds application mimicking native shape-to-function relationship in the context of tissue engineering and provides a platform to further study the roles of micro- and macroscale architectural features that exist within native tissues.


Assuntos
Hidrogéis , Microfluídica , Engenharia Tecidual , Hidrogéis/química , Humanos , Microfluídica/métodos , Microfluídica/instrumentação , Células-Tronco Mesenquimais/citologia
9.
Biomater Sci ; 12(1): 134-150, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-37933486

RESUMO

Synthetic polymers, such as poly(vinyl alcohol) (PVA), are popular biomaterials for the fabrication of hydrogels for tissue engineering and regenerative medicine (TERM) applications, as they provide excellent control over the physico-chemical properties of the hydrogel. However, their bioinert nature is known to limit cell-biomaterial interactions by hindering cell infiltration, blood vessel recruitment and potentially limiting their integration with the host tissue. Efforts in the field have therefore focused on increasing the biofunctionality of synthetic hydrogels, without limiting the advantages associated with their tailorability and controlled release capacity. The aim of this study was to investigate the suitability of pristine gelatin to enhance the biofunctionality of tyraminated PVA (PVA-Tyr) hydrogels, by promoting cell infiltration and host blood vessel recruitment for TERM applications. Pure PVA-Tyr hydrogels and PVA-Tyr hydrogels incorporated with vascular endothelial growth factor (VEGF), a well-known pro-angiogenic stimulus, were used for comparison. Incorporating increasing concentrations of VEGF (0.01-10 µg mL-1) or gelatin (0.01-5 wt%) did not influence the physical properties of PVA-Tyr hydrogels. However, their presence within the polymer network (>0.1 µg mL-1 VEGF and >0.1 wt% gelatin) promoted endothelial cell interactions with the hydrogels. The covalent binding of unmodified gelatin or VEGF to the PVA-Tyr network did not hamper their inherent bioactivity, as they both promoted angiogenesis in a chick chorioallantoic membrane (CAM) assay, performing comparably with the unbound VEGF control. When the PVA-Tyr hydrogels were implanted subcutaneously in mice, it was observed that cell infiltration into the hydrogels was possible in the absence of gelatin or VEGF at 1- or 3-weeks post-implantation, highlighting a clear difference between in vitro an in vivo cell-biomaterial interaction. Nevertheless, the presence of gelatin or VEGF was necessary to enhance blood vessel recruitment and infiltration, although no significant difference was observed between these two biological molecules. Overall, this study highlights the potential of gelatin as a standalone pro-angiogenic cue to enhance biofunctionality of synthetic hydrogels and provides promise for their use in a variety of TERM applications.


Assuntos
Álcool de Polivinil , Fator A de Crescimento do Endotélio Vascular , Camundongos , Animais , Álcool de Polivinil/química , Gelatina/química , Engenharia Tecidual , Hidrogéis/química , Polímeros/química , Materiais Biocompatíveis/farmacologia , Materiais Biocompatíveis/química , Etanol
10.
Adv Healthc Mater ; 12(30): e2300977, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37699146

RESUMO

Volumetric bioprinting (VBP) is a light-based 3D printing platform, which recently prompted a paradigm shift for additive manufacturing (AM) techniques considering its capability to enable the fabrication of complex cell-laden geometries in tens of seconds with high spatiotemporal control and pattern accuracy. A flexible allyl-modified gelatin (gelAGE)-based photoclick resin is developed in this study to fabricate matrices with exceptionally soft polymer networks (0.2-1.0 kPa). The gelAGE-based resin formulations are designed to exploit the fast thiol-ene crosslinking in combination with a four-arm thiolated polyethylene glycol (PEG4SH) in the presence of a photoinitiator. The flexibility of the gelAGE biomaterial platform allows one to tailor its concentration spanning from 2.75% to 6% and to vary the allyl to thiol ratio without hampering the photocrosslinking efficiency. The thiol-ene crosslinking enables the production of viable cell-material constructs with a high throughput in tens of seconds. The suitability of the gelAGE-based resins is demonstrated by adipogenic differentiation of adipose-derived stromal cells (ASC) after VBP and by the printing of more fragile adipocytes as a proof-of-concept. Taken together, this study introduces a soft photoclick resin which paves the way for volumetric printing applications toward soft tissue engineering.


Assuntos
Bioimpressão , Engenharia Tecidual , Engenharia Tecidual/métodos , Gelatina , Bioimpressão/métodos , Hidrogéis , Impressão Tridimensional , Compostos de Sulfidrila , Alicerces Teciduais
11.
Macromol Biosci ; : e2300457, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-38035637

RESUMO

Cell encapsulation within three-dimensional hydrogels is a promising approach to mimic tissues. However, true biomimicry of the intricate microenvironment, biophysical and biochemical gradients, and the macroscale hierarchical spatial organizations of native tissues is an unmet challenge within tissue engineering. This review provides an overview of the macromolecular chemistries that have been applied toward the design of cell-friendly hydrogels, as well as their application toward controlling biophysical and biochemical bulk and gradient properties of the microenvironment. Furthermore, biofabrication technologies provide the opportunity to simultaneously replicate macroscale features of native tissues. Biofabrication strategies are reviewed in detail with a particular focus on the compatibility of these strategies with the current macromolecular toolkit described for hydrogel design and the challenges associated with their clinical translation. This review identifies that the convergence of the ever-expanding macromolecular toolkit and technological advancements within the field of biofabrication, along with an improved biological understanding, represents a promising strategy toward the successful tissue regeneration.

12.
Acta Biomater ; 156: 202-213, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-35413478

RESUMO

A tibial tuberosity advancement (TTA), used to treat lameness in the canine stifle, provides a framework to investigate implant performance within an uneven loading environment due to the dominating patellar tendon. The purpose of this study was to reassess how we design orthopaedic implants in a load-bearing model to investigate potential for improved osseointegration capacity of fully-scaffolded mechanically-matched additive manufactured (AM) implants. While the mechanobiological nature of bone is well known, we have identified a lower limit in the literature where investigation into exceedingly soft scaffolds relative to trabecular bone ceases due to the trade-off in mechanical strength. We developed a finite element model of the sheep stifle to assess the stresses and strains of homogeneous and locally-optimised TTA implant designs. Using additive manufacturing, we printed three different low-stiffness Ti-6Al-4 V TTA implants: 0.8 GPa (Ti1), 0.6 GPa (Ti2) and an optimised design with a 0.3 GPa cortex and 0.1 GPa centre (Ti3), for implantation in a 12-week in vivo ovine pilot study. Static histomorphometry demonstrated uniform bone ingrowth in optimised low-modulus Ti3 samples compared to homogeneous designs (Ti1 and Ti2), and greater bone-implant contact. Mineralising surfaces were apparent in all implants, though mineral apposition rate was only consistent throughout Ti3. The greatest bone formation scores were seen in Ti3, followed by Ti2 and Ti1. Results from our study suggest lower stiffnesses and higher strain ranges improve early bone formation, and that by accounting for loading environments through rational design, implants can be optimised to improve uniform osseointegration. STATEMENT OF SIGNIFICANCE: The effect of different strain ranges on bone healing has been traditionally investigated and characterised through computational models, with much of the literature suggesting higher strain ranges being favourable. However, little has been done to incorporate strain-optimisation into porous orthopaedic implants due to the trade-off in mechanical strength required to induce these microenvironments. In this study, we used finite element analysis to optimise the design of additive manufactured (AM) titanium orthopaedic implants for different strain ranges, using a clinically-relevant surgical model. Our research suggests that there is potential for locally-optimised AM scaffolds in the use of orthopaedic devices to induce higher strains, which in turn encourages de novo bone formation and uniform osseointegration.


Assuntos
Osteogênese , Titânio , Animais , Ovinos , Cães , Titânio/farmacologia , Projetos Piloto , Próteses e Implantes , Osseointegração , Porosidade , Ligas
13.
J Mater Sci Mater Med ; 23(2): 283-91, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22190196

RESUMO

Magnesium (Mg) alloys are being actively investigated as potential load-bearing orthopaedic implant materials due to their biodegradability in vivo. With Mg biomaterials at an early stage in their development, the screening of alloy compositions for their biodegradation rate, and hence biocompatibility, is reliant on cost-effective in vitro methods. The use of a buffer to control pH during in vitro biodegradation is recognised as critically important as this seeks to mimic pH control as it occurs naturally in vivo. The two different types of in vitro buffer system available are based on either (i) zwitterionic organic compounds or (ii) carbonate buffers within a partial-CO(2) atmosphere. This study investigated the influence of the buffering system itself on the in vitro corrosion of Mg. It was found that the less realistic zwitterion-based buffer did not form the same corrosion layers as the carbonate buffer, and was potentially affecting the behaviour of the hydrated oxide layer that forms on Mg in all aqueous environments. Consequently it was recommended that Mg in vitro experiments use the more biorealistic carbonate buffering system when possible.


Assuntos
Magnésio/química , Ligas , Materiais Biocompatíveis/química , Biodegradação Ambiental , Líquidos Corporais , Soluções Tampão , Carbono/química , Corrosão , Análise Custo-Benefício , Meios de Cultura/química , Gases , Humanos , Hidrogênio/química , Concentração de Íons de Hidrogênio , Teste de Materiais , Microscopia Eletrônica de Varredura/métodos , Ortopedia , Plasma/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier/métodos
14.
Adv Mater ; 34(20): e2107759, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35128736

RESUMO

The field of bioprinting has made significant advancements in recent years and allowed for the precise deposition of biomaterials and cells. However, within this field lies a major challenge, which is developing high resolution constructs, with complex architectures. In an effort to overcome these challenges a biofabrication technique known as vat polymerization is being increasingly investigated due to its high fabrication accuracy and control of resolution (µm scale). Despite the progress made in developing hydrogel precursors for bioprinting techniques, such as extrusion-based bioprinting, there is a major lack in developing hydrogel precursor bioresins for vat polymerization. This is due to the specific unique properties and characteristics required for vat polymerization, from lithography to the latest volumetric printing. This is of major concern as the shortage of bioresins available has a significant impact on progressing this technology and exploring its full potential, including speed, resolution, and scale. Therefore, this review discusses the key requirements that need to be addressed in successfully developing a bioresin. The influence of monomer architecture and bioresin composition on printability is described, along with key fundamental parameters that can be altered to increase printing accuracy. Finally, recent advancements in bioresins are discussed together with future directions.


Assuntos
Bioimpressão , Engenharia Tecidual , Bioimpressão/métodos , Hidrogéis , Polimerização , Impressão Tridimensional , Engenharia Tecidual/métodos , Alicerces Teciduais
15.
Trends Biotechnol ; 40(1): 77-92, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34016480

RESUMO

Autologous fat grafting offers significant promise for the repair of soft tissue deformities; however, high resorption rates indicate that engineered solutions are required to improve adipose tissue (AT) survival. Advances in material development and biofabrication have laid the foundation for the generation of functional AT constructs; however, a balance needs to be struck between clinically feasible delivery and improved structural integrity of the grafts. A new approach combining the objectives from both the clinical and research communities will assist in developing morphologically and genetically mature AT constructs, with controlled spatial arrangement and increased potential for neovascularization. In a rapidly progressing field, this review addresses research in both the preclinical and bioengineering domains and assesses their ability to resolve functional challenges.


Assuntos
Tecido Adiposo , Transplante Autólogo
16.
Bone ; 154: 116198, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34534709

RESUMO

Tissue engineering approaches for bone repair have rapidly evolved due to the development of novel biofabrication technologies, providing an opportunity to fabricate anatomically-accurate living implants with precise placement of specific cell types. However, limited availability of biomaterial inks, that can be 3D-printed with high resolution, while providing high structural support and the potential to direct cell differentiation and maturation towards the osteogenic phenotype, remains an ongoing challenge. Aiming towards a multifunctional biomaterial ink with high physical stability and biological functionality, this work describes the development of a nanocomposite biomaterial ink (Mg-PCL) comprising of magnesium hydroxide nanoparticles (Mg) and polycaprolactone (PCL) thermoplastic for 3D printing of strong and bioactive bone regenerative scaffolds. We characterised the Mg nanoparticle system and systematically investigated the cytotoxic and osteogenic effects of Mg supplementation to human mesenchymal stromal cells (hMSCs) 2D-cultures. Next, we prepared Mg-PCL biomaterial ink using a solvent casting method, and studied the effect of Mg over mechanical properties, printability and scaffold degradation. Furthermore, we delivered MSCs within Mg-PCL scaffolds using a gelatin-methacryloyl (GelMA) matrix, and evaluated the effect of Mg over cell viability and osteogenic differentiation. Nanocomposite Mg-PCL could be printed with high fidelity at 20 wt% of Mg content, and generated a mechanical reinforcement between 30%-400% depending on the construct internal geometry. We show that Mg-PCL degrades faster than standard PCL in an accelerated-degradation assay, which has positive implications towards in vivo implant degradation and bone regeneration. Mg-PCL did not affect MSCs viability, but enhanced osteogenic differentiation and bone-specific matrix deposition, as demonstrated by higher ALP/DNA levels and Alizarin Red calcium staining. Finally, we present proof of concept of Mg-PCL being utilised in combination with a bone-specific bioink (Sr-GelMA) in a coordinated-extrusion bioprinting strategy for fabrication of hybrid constructs with high stability and synergistic biological functionality. Mg-PCL further enhanced the osteogenic differentiation of encapsulated MSCs and supported bone ECM deposition within the bioink component of the hybrid construct, evidenced by mineralised nodule formation, osteocalcin (OCN) and collagen type-I (Col I) expression within the bioink filaments. This study demonstrated that magnesium-based nanocomposite bioink material optimised for extrusion-based 3D printing of bone regenerative scaffolds provide enhanced mechanical stability and bone-related bioactivity with promising potential for skeletal tissue regeneration.


Assuntos
Bioimpressão , Nanocompostos , Bioimpressão/métodos , Regeneração Óssea , Nanocompostos/química , Osteogênese , Engenharia Tecidual/métodos , Alicerces Teciduais/química
17.
Biofabrication ; 14(3)2022 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-35320796

RESUMO

Bone regeneration of critical-sized bone defects, bone fractures or joint replacements remains a significant clinical challenge. Although there has been rapid advancement in both the fields of bone tissue engineering and additive manufacturing, functional bone implants with rapid vascularization capacity to ensure osseointegration and long-term biological fixation in large bone defects remains limited in clinics. In this study, we developed anin vitrovascularized bone implant by combining cell-laden hydrogels with direct metal printed (DMP) porous titanium alloys (Ti-6Al-4V). A 5 wt% allylated gelatin (GelAGE), was utilized to co-encapsulate human mesenchymal stromal cells (hMSCs) and human umbilical vein endothelial cells (HUVECs) to investigate concurrent osteogenic and vasculogenic performance. DMP macro-porous Ti-6Al-4V scaffolds were subsequently infused/enriched with cell-laden GelAGE to examine the feasibility to deliver cells and engineer vascular-like networks in the hybrid implant. Furthermore, as a proof of concept, a full-scale porous Ti-6Al-4V acetabular cup was impregnated with cell-laden hydrogel to validate the clinical potential of this strategy. The vasculogenic potential was evaluated by examining micro-capillary formation coupled with capillary network maturation and stabilization. Osteogenic differentiation was assessed via alkaline phosphatase activity as well as osteocalcin and osteopontin expression. Our results suggested that GelAGE supported HUVECs spreading and vascular-like network formation, along with osteogenesis of hMSCs. Titanium hybrid constructs with cell-laden hydrogel demonstrated enhanced osteogenesis with similar vasculogenic capability compared to the cell-laden hydrogel alone constructs. The full-scale implant with cell-laden hydrogel coating similarly showed cell distribution and spreading, implying the potential for further clinical application. Our study presents the feasibility of integrating bio-functional hydrogels with porous titanium implants to fabricate a vascularized hybrid construct with both mechanical support and preferable biological functionality (osteogenesis/vasculogenesis), which paves the way for improved strategies to enhance bone regeneration in complex large bone defects achieving long-term bone-implant fixation.


Assuntos
Osseointegração , Osteogênese , Células Endoteliais , Humanos , Hidrogéis/farmacologia , Titânio/farmacologia
18.
J Funct Biomater ; 13(2)2022 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-35466223

RESUMO

Epigenetic approaches using the histone deacetylase 2 and 3 inhibitor-MI192 have been reported to accelerate stem cells to form mineralised tissues. Gelatine methacryloyl (GelMA) hydrogels provide a favourable microenvironment to facilitate cell delivery and support tissue formation. However, their application for bone repair is limited due to their low mechanical strength. This study aimed to investigate a GelMA hydrogel reinforced with a 3D printed scaffold to support MI192-induced human bone marrow stromal cells (hBMSCs) for bone formation. Cell culture: The GelMA (5 wt%) hydrogel supported the proliferation of MI192-pre-treated hBMSCs. MI192-pre-treated hBMSCs within the GelMA in osteogenic culture significantly increased alkaline phosphatase activity (p ≤ 0.001) compared to control. Histology: The MI192-pre-treated group enhanced osteoblast-related extracellular matrix deposition and mineralisation (p ≤ 0.001) compared to control. Mechanical testing: GelMA hydrogels reinforced with 3D printed poly(ethylene glycol)-terephthalate/poly(butylene terephthalate) (PEGT/PBT) scaffolds exhibited a 1000-fold increase in the compressive modulus compared to the GelMA alone. MI192-pre-treated hBMSCs within the GelMA-PEGT/PBT constructs significantly enhanced extracellular matrix collagen production and mineralisation compared to control (p ≤ 0.001). These findings demonstrate that the GelMA-PEGT/PBT construct provides enhanced mechanical strength and facilitates the delivery of epigenetically-activated MSCs for bone augmentation strategies.

19.
Am J Sports Med ; 50(4): 1044-1053, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35188803

RESUMO

BACKGROUND: The lack of healing at the repaired tendon-bone interface is an important cause of failure after rotator cuff repair. While augmentation with growth factors (GFs) has demonstrated promise, the ideal combination must target all 3 tissue types at the tendon-bone interface. HYPOTHESIS: The GF combination of transforming growth factor beta 1, Insulin-like growth factor 1, and parathyroid hormone will promote tenocyte proliferation and differentiation and improve the biomechanical and histological quality of the repaired tendon-bone interface. STUDY DESIGN: Controlled laboratory study. METHODS: In vitro, human tenocytes were cultured in the presence of the GF combination for 72 hours, and cell growth assays and the expression of genes specific to tendon, cartilage, and bone were analyzed. In vivo, adult rats (N = 46) underwent detachment and repair of the left supraspinatus tendon. A PVA-tyramine gel was used to deliver the GF combination to the tendon-bone interface. Histological, biomechanical, and RNA microarray analysis was performed at 6 and 12 weeks after surgery. Immunohistochemistry for type II and X collagen was performed at 12 weeks. RESULTS: When treated with the GF combination in vitro, human tenocytes proliferated 1.5 times more than control (P = .04). The expression of scleraxis increased 65-fold (P = .013). The expression of Sox-9 (P = .011), type I collagen (P = .021), fibromodulin (P = .0075), and biglycan (P = .010) was also significantly increased, while the expression of PPARγ was decreased (P = .007). At 6 and 12 weeks postoperatively, the quality of healing on histology was significantly higher in the GF group, with the formation of a more mature tendon-bone interface, as confirmed by immunohistochemistry for type II and X collagen. The GF group achieved a load at failure and Young modulus >1.5 times higher at both time points. Microarrays at 6 weeks demonstrated upregulation of genes involved in leukocyte aggregation (S100A8, S100A9) and tissue mineralization (Bglap, serglycin, Fam20c). CONCLUSION: The GF combination promoted protendon and cartilage responses in human tenocytes in vitro; it also improved the histological appearance and mechanical properties of the repair in vivo. Microarrays of the tendon-bone interface identified inflammatory and mineralization pathways affected by the GF combination, providing novel therapeutic targets for further research. CLINICAL RELEVANCE: The use of this GF combination is translatable to patients and may improve healing after rotator cuff repair.


Assuntos
Lesões do Manguito Rotador , Manguito Rotador , Animais , Fenômenos Biomecânicos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico , Ratos , Manguito Rotador/patologia , Lesões do Manguito Rotador/patologia , Lesões do Manguito Rotador/cirurgia , Cicatrização/fisiologia
20.
Adv Healthc Mater ; 11(2): e2101873, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34710291

RESUMO

The principle challenge for engineering viable, cell-laden hydrogel constructs of clinically-relevant size, is rapid vascularization, in order to moderate the finite capacity of passive nutrient diffusion. A multiscale vascular approach, with large open channels and bulk microcapillaries may be an admissible approach to accelerate this process, promoting overall pre-vascularization for long-term viability of constructs. However, the limited availability of bioinks that possess suitable characteristics that support both fabrication of complex architectures and formation of microcapillaries, remains a barrier to advancement in this space. In this study, gelatin-norbornene (Gel-NOR) is investigated as a vascular bioink with tailorable physico-mechanical properties, which promoted the self-assembly of human stromal and endothelial cells into microcapillaries, as well as being compatible with extrusion and lithography-based biofabrication modalities. Gel-NOR constructs containing self-assembled microcapillaries are successfully biofabricated with varying physical architecture (fiber diameter, spacing, and orientation). Both channel sizes and cell types affect the overall structural changes of the printed constructs, where cross-signaling between both human stromal and endothelial cells may be responsible for the reduction in open channel lumen observed over time. Overall, this work highlights an exciting three-way interplay between bioink formulation, construct design, and cell-mediated response that can be exploited towards engineering vascular tissues.


Assuntos
Bioimpressão , Capilares , Gelatina , Engenharia Tecidual , Capilares/crescimento & desenvolvimento , Células Endoteliais , Gelatina/química , Humanos , Hidrogéis/química , Norbornanos/química , Impressão Tridimensional , Alicerces Teciduais/química
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa