Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 43(9): 1639-1652, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37409527

RESUMO

BACKGROUND: Treatment of occluded vessels can involve angioplasty, stenting, and bypass grafting, which can be limited by restenosis and thrombosis. Drug-eluting stents attenuate restenosis, but the current drugs used are cytotoxic, causing smooth muscle cell (SMC) and endothelial cell (EC) death that may lead to late thrombosis. N-cadherin is a junctional protein expressed by SMCs, which promotes directional SMC migration contributing to restenosis. We propose that engaging N-cadherin with mimetic peptides can act as a cell type-selective therapeutic strategy to inhibit polarization and directional migration of SMCs without negatively impacting ECs. METHODS: We designed a novel N-cadherin-targeting chimeric peptide with a histidine-alanine-valine cadherin-binding motif, combined with a fibronectin-binding motif from Staphylococcus aureus. This peptide was tested in SMC and EC culture assays of migration, viability, and apoptosis. Rat carotid arteries were balloon injured and treated with the N-cadherin peptide. RESULTS: Treating scratch-wounded SMCs with the N-cadherin-targeting peptide inhibited migration and reduced polarization of wound-edge cells. The peptide colocalized with fibronectin. Importantly, EC junction, permeability, or migration was not impacted by peptide treatment in vitro. We also demonstrated that the chimeric peptide persisted for 24 hours after transient delivery in the balloon-injured rat carotid artery. Treatment with the N-cadherin-targeting chimeric peptide reduced intimal thickening in balloon-injured rat carotid arteries at 1 and 2 weeks after injury. Reendothelialization of injured vessels after 2 weeks was unimpaired by peptide treatment. CONCLUSIONS: These studies show that an N-cadherin-binding and fibronectin-binding chimeric peptide is effective in inhibiting SMC migration in vitro and in vivo and limiting neointimal hyperplasia after balloon angioplasty without affecting EC repair. These results establish the potential of an advantageous SMC-selective strategy for antirestenosis therapy.


Assuntos
Lesões das Artérias Carótidas , Trombose , Ratos , Animais , Fibronectinas/farmacologia , Lesões das Artérias Carótidas/patologia , Caderinas , Artérias Carótidas/patologia , Hiperplasia/patologia , Peptídeos/farmacologia , Trombose/patologia
2.
Arterioscler Thromb Vasc Biol ; 43(7): 1096-1110, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37226729

RESUMO

Despite recent advancements in vascular disease treatments, thrombosis and poor long-term vessel patency remain significant barriers to effective endovascular intervention. Current balloon angioplasty and stenting techniques effectively restore acute blood flow in occluded vessels but have persistent limitations. Damage to the arterial endothelium caused by injury during catheter tracking triggers neointimal hyperplasia and the release of proinflammatory factors leading to increased risk of thrombosis and restenosis. Antirestenotic agents commonly delivered on angioplasty balloons and stents have lowered arterial restenosis rates, but the absence of cell type selectivity significantly delays critical endothelium repair. Targeted delivery of biomolecular therapeutics, coupled with engineered nanoscale excipients, has the potential to redefine cardiovascular interventions by improving long-term efficacy, limiting off-target effects, and reducing costs compared with conventional clinical standards of care. This review analyzes current forms of localized vascular drug delivery, emerging nanoscale therapeutic and excipient strategies, and provides recommendations for future areas of study to advance the treatment of vascular disease through innovations in nanotechnology.


Assuntos
Angioplastia com Balão , Trombose , Doenças Vasculares , Humanos , Angioplastia com Balão/efeitos adversos , Angioplastia com Balão/métodos , Stents , Constrição Patológica/etiologia , Doenças Vasculares/etiologia , Trombose/etiologia , Nanotecnologia , Resultado do Tratamento
3.
J Magn Reson Imaging ; 58(4): 1139-1150, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-36877190

RESUMO

BACKGROUND: A noninvasive method to track implanted biomaterials is desirable for real-time monitoring of material interactions with host tissues and assessment of efficacy and safety. PURPOSE: To explore quantitative in vivo tracking of polyurethane implants using a manganese porphyrin (MnP) contrast agent containing a covalent binding site for pairing to polymers. STUDY TYPE: Prospective, longitudinal. ANIMAL MODEL: Rodent model of dorsal subcutaneous implants (10 female Sprague Dawley rats). FIELD STRENGTH/SEQUENCE: A 3-T; two-dimensional (2D) T1-weighted spin-echo (SE), T2-weighted turbo SE, three-dimensional (3D) spoiled gradient-echo T1 mapping with variable flip angles. ASSESSMENT: A new MnP-vinyl contrast agent to covalently label polyurethane hydrogels was synthesized and chemically characterized. Stability of binding was assessed in vitro. MRI was performed in vitro on unlabeled hydrogels and hydrogels labeled at different concentrations, and in vivo on rats with unlabeled and labeled hydrogels implanted dorsally. In vivo MRI was performed at 1, 3, 5, and 7 weeks postimplantation. Implants were easily identified on T1-weighted SE, and fluid accumulation from inflammation was distinguished on T2-weighted turbo SE. Implants were segmented on contiguous T1-weighted SPGR slices using a threshold of 1.8 times the background muscle signal intensity; implant volume and mean T1 values were then calculated at each timepoint. Histopathology was performed on implants in the same plane as MRI and compared to imaging results. STATISTICAL TESTS: Unpaired t-tests and one-way analysis of variance (ANOVA) were used for comparisons. A P value <0.05 was considered to be statistically significant. RESULTS: Hydrogel labeling with MnP resulted in a significant T1 reduction in vitro (T1 = 517 ± 36 msec vs. 879 ± 147 msec unlabeled). Mean T1 values of labeled implants in rats increased significantly by 23% over time, from 1 to 7 weeks postimplantation (651 ± 49 msec to 801 ± 72 msec), indicating decreasing implant density. DATA CONCLUSION: Polymer-binding MnP enables in vivo tracking of vinyl-group coupling polymers. EVIDENCE LEVEL: 1. TECHNICAL EFFICACY: Stage 1.


Assuntos
Meios de Contraste , Porfirinas , Feminino , Ratos , Animais , Poliuretanos , Manganês , Hidrogéis , Estudos Prospectivos , Ratos Sprague-Dawley , Imageamento por Ressonância Magnética/métodos
4.
Biomacromolecules ; 24(7): 3149-3158, 2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37344353

RESUMO

In a healthy heart, cells naturally secrete C-type natriuretic peptide (CNP), a cytokine that protects against myofibroblast differentiation of cardiac fibroblasts and extracellular matrix deposition leading to fibrosis. CNP availability during myocardial remodeling is important to prevent cardiac fibrosis, but CNP is limited after an injury because of the loss of cardiomyocytes and the activation of cardiac fibroblasts to myofibroblasts. We hypothesized that the sustained release of exogenous CNP from oligo-urethane nanoparticles (NPs) would reduce differentiation of human cardiac fibroblasts toward a myofibrogenic phenotype. Our work used a modified form of a degradable polar hydrophobic ionic (D-PHI) oligo-urethane, which has shown the ability to self-assemble into NPs for the delivery of peptide and oligonucleotide biomolecules. The CNP-loaded NPs (NPCNP) were characterized for a diameter of 129 ± 1.4 nm and a ζ potential of -46 ± 7.8 mV. Treatment of cardiac fibroblasts with NPCNP increased cyclic guanosine-monophosphate (cGMP) synthesis, confirming that exogenous CNP delivered via oligo-urethane NPs is bioactive and can induce downstream signaling that has been implicated in antagonizing transforming growth factor-ß1 (TGF-ß1)-induced myofibrogenic differentiation. It is also shown that treatment with NPCNP attenuated contraction of collagen gels by cardiac myofibroblasts stimulated with TGF-ß1. Coating with heparin on the NPCNP (HEP-NPCNP) exemplified an approach to extend the release of CNP from the NPs. Both HEP-NPCNP and NPCNP show minimal cell toxicity, studied up to 0.25 × 1010 NPs/mL in culture media. These findings support further investigation of CNP delivery via NPs as a future therapy for suppressing cardiac fibrosis.


Assuntos
Miofibroblastos , Fator de Crescimento Transformador beta1 , Humanos , Peptídeo Natriurético Tipo C/farmacologia , Uretana , Fibrose
5.
Int J Mol Sci ; 23(9)2022 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-35563649

RESUMO

There is a shortage of suitable tissue-engineered solutions for gingival recession, a soft tissue defect of the oral cavity. Autologous tissue grafts lead to an increase in morbidity due to complications at the donor site. Although material substitutes are available on the market, their development is early, and work to produce more functional material substitutes is underway. The latter materials along with newly conceived tissue-engineered substitutes must maintain volumetric form over time and have advantageous mechanical and biological characteristics facilitating the regeneration of functional gingival tissue. This review conveys a comprehensive and timely perspective to provide insight towards future work in the field, by linking the structure (specifically multilayered systems) and function of electrospun material-based approaches for gingival tissue engineering and regeneration. Electrospun material composites are reviewed alongside existing commercial material substitutes', looking at current advantages and disadvantages. The importance of implementing physiologically relevant degradation profiles and mechanical properties into the design of material substitutes is presented and discussed. Further, given that the broader tissue engineering field has moved towards the use of pre-seeded scaffolds, a review of promising cell options, for generating tissue-engineered autologous gingival grafts from electrospun scaffolds is presented and their potential utility and limitations are discussed.


Assuntos
Engenharia Tecidual , Alicerces Teciduais , Alicerces Teciduais/química
6.
Biomacromolecules ; 18(8): 2296-2305, 2017 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-28621927

RESUMO

The use of degradable polymers in vascular tissue regeneration has sparked the need to characterize polymer biocompatibility during degradation. While tissue compatibility has been frequently addressed, studies on polymer hemocompatibility during degradation are limited. The current study evaluated the differences in hemocompatibility (platelet response, complement activation, and coagulation cascade initiation) between as-made and hydrolyzed poly(lactic-co-glycolic) acid (PLGA) and degradable polar hydrophobic ionic polyurethane (D-PHI). Platelet activation decreased (in whole blood) and platelet adhesion decreased (in blood without leukocytes) for degraded versus as-made PLGA. D-PHI showed minimal hemocompatibility changes over degradation. Leukocytes played a major role in mediating platelet activation for samples and controls, as well as influencing platelet-polymer adhesion on the degraded materials. This study demonstrates the importance of assessing the blood compatibility of biomaterials over the course of degradation since the associated changes in surface chemistry and physical state could significantly change biomaterial hemocompatibility.


Assuntos
Plaquetas/metabolismo , Ácido Láctico , Teste de Materiais , Adesividade Plaquetária/efeitos dos fármacos , Ácido Poliglicólico , Poliuretanos , Humanos , Ácido Láctico/química , Ácido Láctico/farmacocinética , Ácido Láctico/farmacologia , Ácido Poliglicólico/química , Ácido Poliglicólico/farmacocinética , Ácido Poliglicólico/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Poliuretanos/química , Poliuretanos/farmacocinética , Poliuretanos/farmacologia
7.
Dent Mater ; 40(10): 1624-1634, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39084955

RESUMO

OBJECTIVE: The biodegradation of methacrylate (MA)-based dental restoratives has been suggested to contribute to a loss of adhesion and subsequent detachment, or secondary caries, both major causes of restoration failure. Previous studies have demonstrated that intermolecular interactions between resin monomers may affect the hydrolytic-susceptibility of composites. Altering the intermolecular interactions by shielding or masking the hydrolytically-susceptible ester groups found in MA monomers could be an effective strategy to mitigate the biodegradation of resin composites. The objective of this work was to assess whether shielding/masking MAs using fluorinated groups could improve the biostability of experimental composites. METHODS: Eight fluorinated monomers (FM) were synthesized, characterized (1H NMR), and formulated into experimental resin composites (FC, 65 wt%, microfill). FCs were assessed for interactions with water (water contact angle, water sorption, gel fraction), mechanical properties (both compressive and flexural strength and modulus), cytocompatibility, resistance to biodegradation using simulated human salivary esterase (SHSE) and compared to a control composite (CC) without FM. RESULTS: Integration of FMs was found to generally decrease both the physical and mechanical properties under all incubation conditions when compared to the CC. Additionally, all FCs had a negative influence on composite biodegradation following immersion in SHSE when compared to the CC. SIGNIFICANCE: Shielding/masking MA-esters inherently inserts molecular spaces between the polymer chains within the resin network, and shielding is likely not possible while also maintaining the necessary cohesive forces that regulate the physical and mechanical properties of resin composites. Novel dental resin development should seek to remove/replace vulnerable ester-containing MAs rather that adopting a shielding/masking approach.


Assuntos
Resinas Compostas , Teste de Materiais , Metacrilatos , Poliuretanos , Resinas Compostas/química , Metacrilatos/química , Hidrólise , Poliuretanos/química , Humanos , Resistência à Flexão , Força Compressiva
8.
Acta Biomater ; 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39303833

RESUMO

Degradable polar hydrophobic ionic polyurethanes (D-PHI) are an emerging class of biomaterials with particular significance for blood-contacting applications due to their immunomodulatory effects and highly customizable block chemistry. In this manuscript, D-PHI polymer was formulated as a nanoparticle excipient for the first time by inverse emulsion polymerization. The nanoparticles were optimized with consideration of diameter, surface charge, size variability, and yield as a delivery vehicle for a custom vascular therapeutic peptide. A layer-by-layer (LBL) surface modification technique using poly-L-lysine was integrated within the nanoparticle design to optimize therapeutic loading efficiency. Solvent pH played a pivotal role in emulsion micelle formation, LBL polymer secondary structure, and the polymer functional group interactions critical for high therapeutic loading. The resulting nanoparticle platform met target size (200 ± 20 nm), polydispersity (<0.07), and storage stability standards, was nontoxic, and did not affect therapeutic peptide bioactivity in vitro. Surface-modified D-PHI nanoparticles can be reproducibly manufactured at low cost, generating a highly customizable excipient platform suitable for delivery of biomolecular therapeutics. These nanoparticles have potential applications in vascular drug delivery via localized infusion, drug eluting stents, and drug-coated angioplasty balloons. STATEMENT OF SIGNIFICANCE: Nanoscale excipients have become critical in the delivery of many therapeutics to enhance drug stability and targeted biodistribution through careful design of nanoparticle composition, surface chemistry, and size. This manuscript describes the development of a nanoparticle excipient derived from an immunomodulatory degradable polar hydrophobic ionic polyurethane, in combination with a layer-by-layer surface modification approach utilizing poly-L-lysine, to transport a mimetic peptide targeting smooth muscle cell migration in vascular disease. The nanoparticle platform draws on the effect of pH to maximize drug loading and tailor particle properties. The low cost and easily reproducible system presents a highly customizable platform that can be adapted for therapeutic delivery across a wide range of clinical indications.

9.
Bioengineering (Basel) ; 11(8)2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39199733

RESUMO

The regenerative capacity of the peripheral nervous system is limited, and peripheral nerve injuries often result in incomplete healing and poor outcomes even after repair. Transection injuries that induce a nerve gap necessitate microsurgical intervention; however, even the current gold standard of repair, autologous nerve graft, frequently results in poor functional recovery. Several interventions have been developed to augment the surgical repair of peripheral nerves, and the application of functional biomaterials, local delivery of bioactive substances, electrical stimulation, and allografts are among the most promising approaches to enhance innate healing across a nerve gap. Biocompatible polymers with optimized degradation rates, topographic features, and other functions provided by their composition have been incorporated into novel nerve conduits (NCs). Many of these allow for the delivery of drugs, neurotrophic factors, and whole cells locally to nerve repair sites, mitigating adverse effects that limit their systemic use. The electrical stimulation of repaired nerves in the perioperative period has shown benefits to healing and recovery in human trials, and novel biomaterials to enhance these effects show promise in preclinical models. The use of acellular nerve allografts (ANAs) circumvents the morbidity of donor nerve harvest necessitated by the use of autografts, and improvements in tissue-processing techniques may allow for more readily available and cost-effective options. Each of these interventions aid in neural regeneration after repair when applied independently, and their differing forms, benefits, and methods of application present ample opportunity for synergistic effects when applied in combination.

10.
Acta Biomater ; 173: 336-350, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37989435

RESUMO

New functional materials for engineering gingival tissue are still in the early stages of development. Materials for such applications must maintain volume and have advantageous mechanical and biological characteristics for tissue regeneration, to be an alternative to autografts, which are the current benchmark of care. In this work, methacrylated gelatin (GelMa) was photocrosslinked with synthetic immunomodulatory methacrylated divinyl urethanes and defined monomers to generate composite scaffolds. Using a factorial design, with the synthetic monomers of a degradable polar/hydrophobic/ionic polyurethane (D-PHI) and GelMa, composite materials were electrospun with polycarbonate urethane (PCNU) and light-cured in-flight. The materials had significantly different relative hydrophilicities, with unique biodegradation profiles associated with specific formulations, thereby providing good guidance to achieving desired mechanical characteristics and scaffold resorption for gingival tissue regeneration. In accelerated esterase/collagenase degradation models, the new materials exhibited an initial rapid weight loss followed by a more gradual rate of degradation. The degradation profile allowed for the early infiltration of human adipose-derived stromal/stem cells, while still enabling the graft's structural integrity to be maintained. In conclusion, the materials provide a promising candidate platform for the regeneration of oral soft tissues, addressing the requirement of viable tissue infiltration while maintaining volume and mechanical integrity. STATEMENT OF SIGNIFICANCE: There is a need for the development of more functional and efficacious materials for the treatment of gingival recession. To address significant limitations in current material formulations, we sought to investigate the development of methacrylated gelatin (GelMa) and oligo-urethane/methacrylate monomer composite materials. A factorial design was used to electrospin four new formulations containing four to five monomers. Synthetic immunomodulatory monomers were crosslinked with GelMa and electrospun with a polycarbonate urethane resulting in unique mechanical properties, and resorption rates which align with the original design criteria for gingival tissue engineering. The materials may have applications in tissue engineering and can be readily manufactured. The findings of this work may help better direct the efforts of tissue engineering and material manufacturing.


Assuntos
Engenharia Tecidual , Alicerces Teciduais , Humanos , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Gelatina/farmacologia , Gelatina/química , Tecido Conjuntivo , Poliuretanos/farmacologia , Poliuretanos/química
11.
Acta Biomater ; 175: 214-225, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38158104

RESUMO

The ex vivo endothelialization of small diameter vascular prostheses can prolong their patency. Here, we demonstrate that heterotypic interactions between human adipose tissue-derived endothelial cells and perivascular cells can be exploited to accelerate the endothelialization of an electrospun ionomeric polyurethane scaffold. The scaffold was used to physically separate endothelial cells from perivascular cells to prevent their diffuse neo-intimal hyperplasia and spontaneous tubulogenesis, yet enable their paracrine cross-talk to accelerate the integration of the endothelial cells into a temporally stable endothelial lining of a continuous, elongated, and aligned morphology. Perivascular cells stimulated endothelial basement membrane protein production and suppressed their angiogenic and inflammatory activation to accelerate this biomimetic morphogenesis of the endothelium. These findings demonstrate the feasibility and underscore the value of exploiting heterotypic interactions between endothelial cells and perivascular cells for the fabrication of an endothelial lining intended for small diameter arterial reconstruction. STATEMENT OF SIGNIFICANCE: Adipose tissue is an abundant, accessible, and uniquely dispensable source of endothelial cells and perivascular cells for vascular tissue engineering. While their spontaneous self-assembly into microvascular networks is routinely exploited for the vascularization of engineered tissues, it threatens the temporal stability of an endothelial lining intended for small diameter arterial reconstruction. Here, we demonstrate that an electrospun polyurethane scaffold can be used to physically separate endothelial cells from perivascular cells to prevent their spontaneous capillary morphogenesis, yet enable their cross-talk to promote the formation of a stable endothelium. Our findings demonstrate the feasibility of engineering an endothelial lining from human adipose tissue, poising it for the rapid ex vivo endothelialization of small diameter vascular prostheses in an autologous, patient-specific manner.


Assuntos
Células Endoteliais , Poliuretanos , Humanos , Poliuretanos/metabolismo , Endotélio , Tecido Adiposo/metabolismo , Engenharia Tecidual , Prótese Vascular
12.
J Plast Reconstr Aesthet Surg ; 91: 276-283, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38432085

RESUMO

BACKGROUND: The study aims to assess the safety and effectiveness of BoneTape™, a new resorbable bone fixation device, using a zygomatic fracture model in rabbits. METHODS: The study followed BoneTape™ samples and control (sham) groups over 2-, 6-, and 12-week periods post-zygomaticomaxillary (ZM) osteotomy and zygomaticofrontal (ZF) disarticulation. The osteotomized segments were analyzed for bone healing, inflammatory response, and tissue healing. µCT imaging and histological analysis were used to examine the axial alignment, offset, and quality of new bone formation. RESULTS: BoneTape™ samples demonstrated enhanced maintenance of the initial intraoperative positioning, reduced axial offset, and better alignment when compared with the control group, enabling stable bone healing under physiological loading conditions. Complete union was observed at 12-weeks in both groups. The BoneTape™ group experienced minimal immune and tissue reactions, classically associated with wound healing, and showed an increased number of giant cells at 6 and 12-weeks. CONCLUSION: BoneTape™ represents a promising advancement in osteosynthesis, demonstrating efficacy in maintaining stable zygomatic reconstruction and eliciting minimal immune response in a rabbit model. This study introduces BoneTape™ as a disruptive solution specifically designed for clinical application in cranio-maxillofacial fracture fixation, with the potential to eliminate the use of over-engineered solutions while offering benefits such as ease of application and fewer biologically disruptive steps.


Assuntos
Fraturas Cranianas , Fraturas Zigomáticas , Animais , Coelhos , Fraturas Zigomáticas/diagnóstico por imagem , Fraturas Zigomáticas/cirurgia , Fixadores Internos , Fixação Interna de Fraturas/métodos , Fraturas Cranianas/diagnóstico por imagem , Fraturas Cranianas/cirurgia , Fixação de Fratura , Placas Ósseas
13.
Acta Biomater ; 166: 167-186, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37207744

RESUMO

Biodegradable hydrogels are growing in demand to enable the delivery of biomolecules (e.g. growth factors) for regenerative medicine. This research investigated the resorption of an oligourethane/polyacrylic acid hydrogel, a biodegradable hydrogel which supports tissue regeneration. The Arrhenius model was used to characterize the resorption of the polymeric gels in relevant in vitro conditions, and the Flory-Rehner equation was used to correlate the volumetric swelling ratio with the extent of degradation. The study found that the swelling rate of the hydrogel follows the Arrhenius model at elevated temperatures, estimating degradation time in saline solution at 37°C to be between 5 and 13 months, serving as a preliminary approximation of degradation in vivo. The degradation products had low cytotoxicity towards endothelial cells, and the hydrogel supported stromal cell proliferation. Additionally, the hydrogels were able to release growth factors and maintain the biomolecules' bioactivity towards cell proliferation. The study of the vascular endothelial growth factor (VEGF) release from the hydrogel used a diffusion process model, showing that the electrostatic attraction between VEGF and the anionic hydrogel allowed for controlled and sustained VEGF release over three weeks. In a rat subcutaneous implant model, a selected hydrogel with desired degradation rates exhibited minimal foreign body response and supported M2a macrophage phenotype, and vascularization. The low M1 and high M2a macrophage phenotypes within the implants were associated with tissue integration. This research supports the use of oligourethane/polyacrylic acid hydrogels as a promising material for delivering growth factors and supporting tissue regeneration. STATEMENT OF SIGNIFICANCE: There is a need for degradable elastomeric hydrogels that can support the formation of soft tissues and minimize long-term foreign body responses. An Arrhenius model was used to estimate the relative breakdown of hydrogels, in-vitro. The results demonstrate that hydrogels made from a combination of poly(acrylic acid) and oligo-urethane diacrylates can be designed to resorb over defined periods ranging from months to years depending on the chemical formulation prescribed by the model. The hydrogel formulations also provided for different release profiles of growth factors, relevant to tissue regeneration. In-vivo, these hydrogels had minimal inflammatory effects and showed evidence of integration into the surrounding tissue. The hydrogel approach can help the field design a broader range of biomaterials for tissue regeneration.


Assuntos
Hidrogéis , Fator A de Crescimento do Endotélio Vascular , Ratos , Animais , Hidrogéis/química , Fator A de Crescimento do Endotélio Vascular/metabolismo , Células Endoteliais/metabolismo , Materiais Biocompatíveis/química , Proliferação de Células
14.
Adv Healthc Mater ; 12(17): e2203168, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36849128

RESUMO

Myocardial fibrosis, resulting from myocardial infarction (MI), significantly alters cardiac electrophysiological properties. As fibrotic scar tissue forms, its resistance to incoming action potentials increases, leading to cardiac arrhythmia, and eventually sudden cardiac death or heart failure. Biomaterials are gaining increasing attention as an approach for addressing post-MI arrhythmias. The current study investigates the hypothesis that a bio-conductive epicardial patch can electrically synchronize isolated cardiomyocytes in vitro and rescue arrhythmic hearts in vivo. A new conceived biocompatible, conductive, and elastic polyurethane composite bio-membrane, referred to as polypyrrole-polycarbonate polyurethane (PPy-PCNU), is developed, in which solid-state conductive PPy nanoparticles are distributed throughout an electrospun aliphatic PCNU nanofiber patch in a controlled manner. Compared to PCNU alone, the resulting biocompatible patch demonstrates up to six times less impedance, with no conductivity loss over time, as well as being able to influence cellular alignment. Furthermore, PPy-PCNU promotes synchronous contraction of isolated neonatal rat cardiomyocytes and alleviates atrial fibrillation in rat hearts upon epicardial implantation. Taken together, epicardially-implanted PPy-PCNU could potentially serve as a novel alternative approach for the treatment of cardiac arrhythmias.


Assuntos
Infarto do Miocárdio , Polímeros , Ratos , Animais , Poliuretanos , Elastômeros , Pirróis/farmacologia , Miócitos Cardíacos , Infarto do Miocárdio/terapia , Arritmias Cardíacas , Condutividade Elétrica
15.
Stem Cell Res Ther ; 14(1): 96, 2023 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-37076906

RESUMO

BACKGROUND: Constructs currently used to repair or replace congenitally diseased pediatric heart valves lack a viable cell population capable of functional adaptation in situ, necessitating repeated surgical intervention. Heart valve tissue engineering (HVTE) can address these limitations by producing functional living tissue in vitro that holds the potential for somatic growth and remodelling upon implantation. However, clinical translation of HVTE strategies requires an appropriate source of autologous cells that can be non-invasively harvested from mesenchymal stem cell (MSC)-rich tissues and cultured under serum- and xeno-free conditions. To this end, we evaluated human umbilical cord perivascular cells (hUCPVCs) as a promising cell source for in vitro production of engineered heart valve tissue. METHODS: The proliferative, clonogenic, multilineage differentiation, and extracellular matrix (ECM) synthesis capacities of hUCPVCs were evaluated in a commercial serum- and xeno-free culture medium (StemMACS™) on tissue culture polystyrene and benchmarked to adult bone marrow-derived MSCs (BMMSCs). Additionally, the ECM synthesis potential of hUCPVCs was evaluated when cultured on polycarbonate polyurethane anisotropic electrospun scaffolds, a representative biomaterial for in vitro HVTE. RESULTS: hUCPVCs had greater proliferative and clonogenic potential than BMMSCs in StemMACS™ (p < 0.05), without differentiation to osteogenic and adipogenic phenotypes associated with valve pathology. Furthermore, hUCPVCs cultured with StemMACS™ on tissue culture plastic for 14 days synthesized significantly more total collagen, elastin, and sulphated glycosaminoglycans (p < 0.05), the ECM constituents of the native valve, than BMMSCs. Finally, hUCPVCs retained their ECM synthesizing capacity after 14 and 21 days in culture on anisotropic electrospun scaffolds. CONCLUSION: Overall, our findings establish an in vitro culture platform that uses hUCPVCs as a readily-available and non-invasively sourced autologous cell population and a commercial serum- and xeno-free culture medium to increase the translational potential of future pediatric HVTE strategies. This study evaluated the proliferative, differentiation and extracellular matrix (ECM) synthesis capacities of human umbilical cord perivascular cells (hUCPVCs) when cultured in serum- and xeno-free media (SFM) against conventionally used bone marrow-derived MSCs (BMMSCs) and serum-containing media (SCM). Our findings support the use of hUCPVCs and SFM for in vitro heart valve tissue engineering (HVTE) of autologous pediatric valve tissue. Figure created with BioRender.com.


Assuntos
Células-Tronco Mesenquimais , Engenharia Tecidual , Adulto , Humanos , Criança , Cordão Umbilical , Diferenciação Celular , Meios de Cultura , Células Cultivadas , Proliferação de Células
16.
STAR Protoc ; 4(1): 101933, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36574341

RESUMO

Here, we describe a protocol for purifying functional clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) from Staphylococcus aureus within 24 h and over 90% purity. SaCas9 purification begins with immobilized metal affinity chromatography, followed by cation exchange chromatography, and ended with centrifugal concentrators. The simplicity, cost-effectiveness, and reproducibility of such protocols will enable general labs to produce a sizable amount of Cas9 proteins, further accelerating CRISPR research.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Staphylococcus aureus/genética , Análise Custo-Benefício , Reprodutibilidade dos Testes
17.
Dent Mater ; 38(12): 1827-1840, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36210221

RESUMO

OBJECTIVE: The objective of this review article is to summarize the current literature on dental resin-based restorative (RBR) materials specifically from the perspective of emerging resin technologies, and to provide researchers with structured design criteria enabling the effective screening of new RBR developments. METHODS: The continued failure of newly introduced RBRs to address biostability without compromising function, over the last decade, are presented as a rationale to support different resin-based concepts. Several developments in the field, aimed at addressing the issues facing modern resin-based systems are summarized and their limitations discussed. A design workflow is proposed for evaluating new RBR, considering resource needs. RESULTS: While several alternative resin chemistries have been suggested over the past decade, all have shown serious limitations in replacing MA-based materials, including their limited physical and mechanical properties, and curing kinetics. Additionally, a broad and inconsistent range of laboratory methods have been used to validate these developments, leading to results that are difficult to compare across studies. A design workflow was conceptualized to facilitate the screening of novel RBRs from both a clinical and research perspective. SIGNIFICANCE: While several alternative chemistries have shown some degree of potential in emulating material property aspects of MA-based resins, a complete restorative system that is resistant to biochemical reactions in saliva has yet to achieve broad clinical adoption. To further spur development, it would be useful to have a more systematic design workflow, that may be used to easily screen new resin technologies effectively early in the design phase, so as to mitigate potential performance failures in the clinic.


Assuntos
Resinas Compostas , Restauração Dentária Permanente , Resinas Compostas/química , Restauração Dentária Permanente/métodos , Ésteres , Teste de Materiais
18.
Biomater Biosyst ; 6: 100049, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36824164

RESUMO

Adipose tissue is an abundant, accessible, and uniquely dispensable source of cells for vascular tissue engineering. Despite its intrinsic endothelial cells, considerable effort is directed at deriving endothelium from its resident stem and progenitor cells. Here, we investigate the composition of human adipose tissue and characterize the phenotypes of its constituent cells in order to help ascertain their potential utility for vascular tissue engineering. Unsupervised clustering based on cell-surface protein signatures failed to detect CD45-CD31-VEGFR2+ endothelial progenitor cells within adipose tissue, but supported further investigation of its resident CD45-CD31+ microvascular endothelial cells (HAMVECs) and CD45-CD31- stromal/stem cells (ASCs). The endothelial differentiation of ASCs altered their proteome, but it remained distinct from that of primary endothelial cell controls - as well as HAMVECs - regardless of their arterial-venous specification or macrovascular-microvascular origin. Rather, ASCs retained a proteome indicative of a perivascular phenotype, which was supported by their ability to facilitate the capillary morphogenesis of HAMVECs. This study supports the use of HAMVECs for the generation of endothelium. It suggests that the utility of ASCs for vascular tissue engineering lies in their capacity to remodel the extracellular matrix and to function as mural cells.

19.
J Biomed Mater Res A ; 110(12): 1932-1943, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35851742

RESUMO

Synthetic scaffolds are needed for generating organized neo-myocardium constructs to promote functional tissue repair. This study investigated the biocompatibility of an elastomeric electrospun degradable polar/hydrophobic/ionic polyurethane (D-PHI) composite scaffold with human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). The composite material was electrospun to generate scaffolds, with nanofibres oriented in aligned or random directions. These features enabled the authors to evaluate the effect of characteristic elements which mimic that of the native extracellular matrix (alignment, chemical heterogeneity, and fiber topography) on hiPSC-CMs activity. The functional nature of the hiPSC-CM cultured on gelatin and Matrigel-coated scaffolds were assessed, investigating the influence of protein interactions with the synthetic substrate on subsequent cell phenotype. After 7 days of culture, high hiPSC-CM viability was observed on the scaffolds. The cells on the aligned scaffold were elongated and demonstrated aligned sarcomeres that oriented parallel to the direction of the fibers, while the cells on random scaffolds and a tissue culture polystyrene (TCPS) control did not exhibit such an organized morphology. The hiPSC-CMs cultured on the scaffolds and TCPS expressed similar levels of cardiac troponin-T, but there was a higher expression of ventricular myosin light chain-2 on the D-PHI composite scaffolds versus TCPS, indicating a higher proportion of hiPSC-CM exhibiting a ventricular cardiomyocyte like phenotype. Within 7 days, the hiPSC-CMs on aligned scaffolds and TCPS beat synchronously and had similar conductive velocities. These preliminary results show that aligned D-PHI elastomeric scaffolds allow hiPSC-CMs to demonstrate important cardiomyocytes characteristics, critical to enabling their future potential use for cardiac tissue regeneration.


Assuntos
Células-Tronco Pluripotentes Induzidas , Nanofibras , Humanos , Células Cultivadas , Gelatina/metabolismo , Miócitos Cardíacos , Cadeias Leves de Miosina/metabolismo , Poliestirenos , Poliuretanos , Alicerces Teciduais , Troponina T/metabolismo
20.
Dent J (Basel) ; 10(12)2022 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-36547039

RESUMO

Here, we explored the role of S. mutans's whole cell and discrete fractions in the degradation of type I collagen and dentinal collagen. Type I collagen gels and human demineralized dentin slabs (DS) were incubated in media alone or with one of the following: overnight (O/N) or newly inoculated (NEW) cultures of S. mutans UA159; intracellular proteins, supernatant or bacterial membranes of O/N cultures. Media from all groups were analyzed for protease-mediated release of the collagen-specific imino acid hydroxyproline. Images of type I collagen and DS were analyzed, respectively. Type I collagen degradation was highest for the supernatant (p < 0.05) fractions, followed by intracellular components and O/N cultures. Collagen degradation for DS samples was highest for O/N samples, followed by supernatant, and intracellular components (p < 0.05). There was lower detectable degradation for both type I collagen and DS from NEW culture samples (p < 0.05), and there was no type I collagen or DS degradation detected for bacterial membrane samples. Structural changes to type I collagen gel and dentinal collagen were observed, respectively, following incubation with S. mutans cultures (O/N and NEW), intracellular components, and supernatant. This study demonstrates that intracellular and extracellular proteolytic activities from S. mutans enable this cariogenic bacterium to degrade type I and dentinal collagen in a growth-phase dependent manner, potentially contributing to the progression of dental caries.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA