Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mater Sci Eng C Mater Biol Appl ; 131: 112515, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34857294

RESUMO

Efficient wound treatments to target specific events in the healing process of chronic wounds constitute a significant aim in regenerative medicine. In this sense, nanomedicine can offer new opportunities to improve the effectiveness of existing wound therapies. The aim of this study was to develop catechol bearing polymeric nanoparticles (NPs) and to evaluate their potential in the field of wound healing. Thus, NPs wound healing promoting activities, potential for drug encapsulation and controlled release, and further incorporation in a hydrogel bioink formulation to fabricate cell-laden 3D scaffolds are studied. NPs with 2 and 29 M % catechol contents (named NP2 and NP29) were obtained by nanoprecipitation and presented hydrodynamic diameters of 100 and 75 nm respectively. These nanocarriers encapsulated the hydrophobic compound coumarin-6 with 70% encapsulation efficiency values. In cell culture studies, the NPs had a protective effect in RAW 264.7 macrophages against oxidative stress damage induced by radical oxygen species (ROS). They also presented a regulatory effect on the inflammatory response of stimulated macrophages and promoted upregulation of the vascular endothelial growth factor (VEGF) in fibroblasts and endothelial cells. In particular, NP29 were used in a hydrogel bioink formulation using carboxymethyl chitosan and hyaluronic acid as polymeric matrices. Using a reactive mixing bioprinting approach, NP-loaded hydrogel scaffolds with good structural integrity, shape fidelity and homogeneous NPs dispersion, were obtained. The in vitro catechol NPs release profile of the printed scaffolds revealed a sustained delivery. The bioprinted scaffolds supported viability and proliferation of encapsulated L929 fibroblasts over 14 days. We envision that the catechol functionalized NPs and resulting bioactive bioink presented in this work offer promising advantages for wound healing applications, as they: 1) support controlled release of bioactive catechol NPs to the wound site; 2) can incorporate additional therapeutic functions by co-encapsulating drugs; 3) can be printed into 3D scaffolds with tailored geometries based on patient requirements.


Assuntos
Bioimpressão , Nanopartículas , Catecóis , Células Endoteliais , Humanos , Impressão Tridimensional , Fator A de Crescimento do Endotélio Vascular
2.
Mater Sci Eng C Mater Biol Appl ; 124: 112024, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33947532

RESUMO

Cytotoxic chemotherapy continues to be the main therapeutic option for patients with metastatic breast cancer. Several studies have reported a significant association between chronic inflammation, carcinogenesis and the presence of cancer stem cells (CSC). We hypothesized that the use of non-steroidal anti-inflammatory drugs targeted to the CSC population could help reducing tumor progression and dissemination in otherwise hard to treat metastatic breast cancer. Within this study cationic naproxen (NAP)-bearing polymeric nanoparticles (NPs) were obtained by self-assembly and they were coated with hyaluronic acid (HA) via electrostatic interaction. HA-coated and uncoated NAP-bearing NPs with different sizes were produced by changing the ionic strength of the aqueous preparation solutions (i.e. 300 and 350 nm or 100 and 130 nm in diameter, respectively). HA-NPs were fully characterized in terms of physicochemical parameters and biological response in cancer cells, macrophages and endothelial cells. Our results revealed that HA-coating of NPs provided a better control in NAP release and improved their hemocompatibility, while ensuring a strong CSC-targeting in MCF-7 breast cancer cells. Furthermore, the best polymeric NPs formulation significantly (p < 0.001) reduced MCF-7 cells viability when compared to free drug (i.e. 45 ± 6% for S-HA-NPs and 87 ± 10% for free NAP) by p53-dependent induction of apoptosis; and the migration of these cell line was also significantly (p < 0.01) reduced by the nano-formulated NAP (i.e. 76.4% of open wound for S-HA-NPs and 61.6% of open wound for NAP). This increased anti-cancer activity of HA-NAP-NPs might be related to the induction of apoptosis through alterations of the GSK-3ß-related COX-independent pathway. Overall, these findings suggest that the HA-NAP-NPs have the potential to improve the treatment of advanced breast cancer by increasing the anti-proliferative effect of NAP within the CSC subpopulation.


Assuntos
Antineoplásicos , Neoplasias da Mama , Nanopartículas , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Células Endoteliais , Glicogênio Sintase Quinase 3 beta , Humanos , Receptores de Hialuronatos , Ácido Hialurônico , Naproxeno/farmacologia , Células-Tronco Neoplásicas
3.
Mater Sci Eng C Mater Biol Appl ; 120: 111716, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33545868

RESUMO

Human mesenchymal stem cells (hMSCs) are an attractive source for cell therapies because of their multiple beneficial properties, i.e. via immunomodulation and secretory factors. Microfluidics is particularly attractive for cell encapsulation since it provides a rapid and reproducible methodology for microgel generation of controlled size and simultaneous cell encapsulation. Here, we report the fabrication of hMSC-laden microcarriers based on in situ ionotropic gelation of water-soluble chitosan in a microfluidic device using a combination of an antioxidant glycerylphytate (G1Phy) compound and tripolyphosphate (TPP) as ionic crosslinkers (G1Phy:TPP-microgels). These microgels showed homogeneous size distributions providing an average diameter of 104 ± 12 µm, somewhat lower than that of control (127 ± 16 µm, TPP-microgels). The presence of G1Phy in microgels maintained cell viability over time and upregulated paracrine factor secretion under adverse conditions compared to control TPP-microgels. Encapsulated hMSCs in G1Phy:TPP-microgels were delivered to the subcutaneous space of immunocompromised mice via injection, and the delivery process was as simple as the injection of unencapsulated cells. Immediately post-injection, equivalent signal intensities were observed between luciferase-expressing microgel-encapsulated and unencapsulated hMSCs, demonstrating no adverse effects of the microcarrier on initial cell survival. Cell persistence, inferred by bioluminescence signal, decreased exponentially over time showing relatively higher half-life values for G1Phy:TPP-microgels compared to TPP-microgels and unencapsulated cells. In overall, results position the microfluidics generated G1Phy:TPP-microgels as a promising microcarrier for supporting hMSC survival and reparative activities.


Assuntos
Quitosana , Células-Tronco Mesenquimais , Microgéis , Animais , Sobrevivência Celular , Humanos , Camundongos , Microfluídica
4.
Eur J Med Chem ; 212: 113152, 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33453601

RESUMO

The development of new drugs for musculoskeletal regeneration purposes has attracted much attention in the last decades. In this work, we present three novel vitamin B9 (folic acid)-derivatives bearing divalent cations (ZnFO, MgFO and MnFO), providing their synthesis mechanism and physicochemical characterization. In addition, a strong emphasis has been placed on evaluating their biological properties (along with our previously reported SrFO) using human mesenchymal stem cells (hMSC). In all the cases, pure folate derivatives (MFOs) with a bidentate coordination mode between the metal and the folate anion, and a 1:1 stoichiometry, were obtained in high yields. A non-cytotoxic dose of all the MFOs (50 µg/mL) was demonstrated to modulate by their own the mRNA profiles towards osteogenic-like or fibrocartilaginous-like phenotypes in basal conditions. Moreover, ZnFO increased the alkaline phosphatase activity in basal conditions, while both ZnFO and MnFO increased the matrix mineralization degree in osteoinductive conditions. Thus, we have demonstrated the bioactivity of these novel compounds and the suitability to further studied them in vivo for musculoskeletal regeneration applications.


Assuntos
Materiais Biocompatíveis/química , Ácido Fólico/química , Células-Tronco Mesenquimais/citologia , Sistema Musculoesquelético/citologia , Engenharia Tecidual , Materiais Biocompatíveis/síntese química , Cátions/síntese química , Cátions/química , Células Cultivadas , Portadores de Fármacos/síntese química , Portadores de Fármacos/química , Ácido Fólico/síntese química , Humanos
5.
Carbohydr Polym ; 241: 116269, 2020 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-32507162

RESUMO

Chitosan-based membranes are promising systems for guided bone regeneration. In this work, we used glycerylphytate as ionic crosslinker and osteinductor compound for the fabrication of chitosan membranes as supports for human mesenchymal stem cells. Three different glycerylphytate-crosslinked membranes were developed by changing the crosslinker concentration, from 2.5-10 wt-%, respect to chitosan. Physico-chemical characterization in terms of composition, morphology, and thermal behavior was further analyzed. Swelling degree, crosslinking density, and crosslinker release showed a glycerylphytate content-dependent behavior. Glycerylphytate suggested to improve osteointegration ability of chitosan surfaces by the formation of apatite-like aggregates after incubation in body simulated fluid. Stem cells cultured on the membranes increased their viability over time, and the incorporation of glycerylphytate improved osteogenic and osteoinductivity potential of chitosan by increasing calcium deposition and alkaline phosphatase (ALP) activity on cultured stem cells. These results demonstrated a potential application of glycerylphytate-crosslinked chitosan systems for promising bone tissue regeneration.


Assuntos
Regeneração Óssea , Quitosana/química , Reagentes de Ligações Cruzadas/química , Ácido Fítico/análogos & derivados , Ácido Fítico/química , Fosfatase Alcalina/metabolismo , Adesão Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Humanos , Membranas Artificiais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Engenharia Tecidual
6.
Macromol Biosci ; 20(7): e2000002, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32421256

RESUMO

Recent studies have demonstrated in vivo synergistic immunosuppressive and anti-inflammatory capacity of dexamethasone (Dx) and naproxen (NAP) in collagen-induced arthritis (CIA) rats. However, the molecular basis of this synergistic effect is barely understood. The low solubility of these drugs and their adverse effects hamper their efficacy on the treatment of inflammatory processes making nanoparticulated systems promising candidates to overcome these drawbacks. The aim of this work is the preparation of polymeric nanoparticles (NPs) that combine NAP and Dx in different concentrations, and the evaluation of the expression of key genes related to autoimmune diseases like CIA. To do so, self-assembled polymeric NPs that incorporate covalently-linked NAP and physically entrapped Dx are designed to have hydrodynamic properties that, according to bibliography, may improve retention and colocalization of both drugs at inflammation sites. The rapid uptake of NPs by macrophages is demonstrated using coumarine-6-loaded NPs. Dx is efficiently encapsulated and in vitro biological studies demonstrate that the Dx-loaded NAP-bearing NPs are noncytotoxic and reduce lipopolysaccharide-induced NO released levels at any of the tested concentrations. Moreover, at the molecular level, a significant synergistic reduction of Il12b transcript gene expression when combining Dx and NAP is demonstrated.


Assuntos
Dexametasona/farmacologia , Macrófagos/metabolismo , Nanopartículas/química , Naproxeno/farmacologia , Polímeros/química , Inibidores da Angiogênese/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Morte Celular/efeitos dos fármacos , Polaridade Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Hidrodinâmica , Interações Hidrofóbicas e Hidrofílicas , Subunidade p40 da Interleucina-12 , Macrófagos/efeitos dos fármacos , Camundongos , Peso Molecular , Nanopartículas/toxicidade , Nanopartículas/ultraestrutura , Naproxeno/síntese química , Naproxeno/química , Óxido Nítrico/metabolismo , Espectroscopia de Prótons por Ressonância Magnética , Células RAW 264.7
7.
Mater Sci Eng C Mater Biol Appl ; 105: 110040, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31546368

RESUMO

Chronic wounds are particularly difficult to heal and constitute an important global health care problem. Some key factors that make chronic wounds challenging to heal are attributed to the incessant release of free radicals, which activate the inflammatory system and impair the repair of the wound. Intrinsic characteristics of hydrogels are beneficial for wound healing, but the effective control of free radical levels in the wound and subsequent inflammation is still a challenge. Catechol, the key molecule responsible for the mechanism of adhesion of mussels, has been proven to be an excellent radical scavenger and anti-inflammatory agent. Our approach in this work lies in the preparation of a hybrid system combining the beneficial properties of hydrogels and catechol for its application as a bioactive wound dressing to assist in the treatment of chronic wounds. The hydrogel backbone is obtained through a self-covalent crosslinking between chitosan (Ch) and oxidized hyaluronic acid (HAox) in the presence of a synthetic catechol terpolymer, which is subsequently coordinated to Fe to obtain an interpenetrated polymer network (IPN). The structural analysis, catechol release profiles, in vitro biological behavior and in vivo performance of the IPN are analyzed and compared with the semi-IPN (without Fe) and the Ch/HAox crosslinked hydrogels as controls. Catechol-containing hydrogels present high tissue adhesion strength under wet conditions, support growth, migration and proliferation of hBMSCs, protect cells against oxidative stress damage induce by ROS, and promote down-regulation of the pro-inflammatory cytokine IL-1ß. Furthermore, in vivo experiments reveal their biocompatibility and stability, and histological studies indicate normal inflammatory responses and faster vascularization, highlighting the performance of the IPN system. The novel IPN design also allows for the in situ controlled and sustained delivery of catechol. Therefore, the developed IPN is a suitable ECM-mimic platform with high cell affinity and bioactive functionalities that, together with the controlled catechol release, will enhance the tissue regeneration process and has a great potential for its application as wound dressing.


Assuntos
Adesivos/química , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Catecóis/farmacologia , Hidrogéis/química , Animais , Bovinos , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quitosana/química , Colágeno/metabolismo , Preparações de Ação Retardada , Humanos , Ácido Hialurônico/química , Interleucina-1beta/metabolismo , Ferro/química , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Oxirredução , Polímeros/química , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Suínos
8.
Sci Rep ; 9(1): 11491, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31391524

RESUMO

Phytic acid (PA) is a natural-occurring antioxidant, which plays an important role in many biological processes. PA is recognized as a potent inhibitor of lipid peroxidation because of its high affinity to multivalent cations, and it can play a role in osteogenic processes. However, its powerful chelating capacity is controversial because it can lead to a severe reduction of mineral availability in the organism. For this reason, compounds with beneficial biological properties of PA, but a modular ion binding capacity, are of high interest. In this work, we report the synthesis and physicochemical characterization of two hydroxylic derivatives of PA, named glycerylphytates (GPhy), through a condensation reaction of PA with glycerol (G). Both derivatives present antioxidant properties, measured by ferrozine/FeCl2 method and chelating activity with calcium ions depending on the content of glyceryl groups incorporated. Besides, the hydroxylic modification not only modulates the ion binding affinity of derivatives but also improves their cytocompatibility in human bone marrow mesenchymal cells (MSCs). Furthermore, GPhy derivatives display osteogenic properties, confirmed by COL1A and ALPL expression depending on composition. These positive features convert GPhy compounds into potent alternatives for those skeletal diseases treatments where PA is tentatively applied.


Assuntos
Antioxidantes/farmacologia , Quelantes/farmacologia , Glicerol/farmacologia , Osteogênese/efeitos dos fármacos , Ácido Fítico/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Antioxidantes/química , Cálcio/metabolismo , Cátions Bivalentes/metabolismo , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quelantes/química , Colágeno Tipo I/metabolismo , Compostos Ferrosos/metabolismo , Ferrozina/farmacologia , Glicerol/química , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , Camundongos , Ácido Fítico/análogos & derivados , Ácido Fítico/química , Cultura Primária de Células , Células RAW 264.7 , Testes de Toxicidade Subaguda
9.
Int J Mol Sci ; 20(12)2019 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-31208091

RESUMO

Acrylic bone cements (ABCs) have played a key role in orthopedic surgery mainly in arthroplasties, but their use is increasingly extending to other applications, such as remodeling of cancerous bones, cranioplasties, and vertebroplasties. However, these materials present some limitations related to their inert behavior and the risk of infection after implantation, which leads to a lack of attachment and makes necessary new surgical interventions. In this research, the physicochemical, thermal, mechanical, and biological properties of ABCs modified with chitosan (CS) and graphene oxide (GO) were studied. Fourier transform infrared (FTIR) spectroscopy, proton nuclear magnetic resonance (1H-NMR) scanning electron microscopy (SEM), Raman mapping, thermogravimetric analysis (TGA), differential scanning calorimetry (DSC), compression resistance, mechanical dynamic analysis (DMA), hydrolytic degradation, cell viability, alkaline phosphatase (ALP) activity with human osteoblasts (HOb), and antibacterial activity against Gram-negative bacteria Escherichia coli were used to characterize the ABCs. The results revealed good dispersion of GO nanosheets in the ABCs. GO provided an increase in antibacterial activity, roughness, and flexural behavior, while CS generated porosity, increased the rate of degradation, and decreased compression properties. All ABCs were not cytotoxic and support good cell viability of HOb. The novel formulation of ABCs containing GO and CS simultaneously, increased the thermal stability, flexural modulus, antibacterial behavior, and osteogenic activity, which gives it a high potential for its uses in orthopedic applications.


Assuntos
Antibacterianos , Materiais Biocompatíveis , Cimentos Ósseos , Quitosana , Grafite , Nanocompostos , Antibacterianos/química , Antibacterianos/farmacologia , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Cimentos Ósseos/química , Cimentos Ósseos/farmacologia , Sobrevivência Celular , Quitosana/química , Grafite/química , Humanos , Fenômenos Mecânicos , Microscopia de Força Atômica , Nanocompostos/química , Nanocompostos/ultraestrutura , Espectroscopia de Infravermelho com Transformada de Fourier , Termogravimetria , Difração de Raios X
10.
ACS Appl Mater Interfaces ; 11(4): 3781-3799, 2019 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-30609898

RESUMO

Osteochondral (OC) regeneration faces several limitations in orthopedic surgery, owing to the complexity of the OC tissue that simultaneously entails the restoration of articular cartilage and subchondral bone diseases. In this study, novel biofunctional hierarchical scaffolds composed of a horseradish peroxidase (HRP)-cross-linked silk fibroin (SF) cartilage-like layer (HRP-SF layer) fully integrated into a HRP-SF/ZnSr-doped ß-tricalcium phosphate (ß-TCP) subchondral bone-like layer (HRP-SF/dTCP layer) were proposed as a promising strategy for OC tissue regeneration. For comparative purposes, a similar bilayered structure produced with no ion incorporation (HRP-SF/TCP layer) was used. A homogeneous porosity distribution was achieved throughout the scaffolds, as shown by micro-computed tomography analysis. The ion-doped bilayered scaffolds presented a wet compressive modulus (226.56 ± 60.34 kPa) and dynamic mechanical properties (ranging from 403.56 ± 111.62 to 593.56 ± 206.90 kPa) superior to that of the control bilayered scaffolds (189.18 ± 90.80 kPa and ranging from 262.72 ± 59.92 to 347.68 ± 93.37 kPa, respectively). Apatite crystal formation, after immersion in simulated body fluid (SBF), was observed in the subchondral bone-like layers for the scaffolds incorporating TCP powders. Human osteoblasts (hOBs) and human articular chondrocytes (hACs) were co-cultured onto the bilayered structures and monocultured in the respective cartilage and subchondral bone half of the partitioned scaffolds. Both cell types showed good adhesion and proliferation in the scaffold compartments, as well as adequate integration of the interface regions. Osteoblasts produced a mineralized extracellular matrix (ECM) in the subchondral bone-like layers, and chondrocytes showed GAG deposition. The gene expression profile was different in the distinct zones of the bilayered constructs, and the intermediate regions showed pre-hypertrophic chondrocyte gene expression, especially on the BdTCP constructs. Immunofluorescence analysis supported these observations. This study showed that the proposed bilayered scaffolds allowed a specific stimulation of the chondrogenic and osteogenic cells in the co-culture system together with the formation of an osteochondral-like tissue interface. Hence, the structural adaptability, suitable mechanical properties, and biological performance of the hierarchical scaffolds make these constructs a desired strategy for OC defect regeneration.


Assuntos
Alicerces Teciduais/química , Animais , Fosfatos de Cálcio/química , Condrócitos/fisiologia , Condrogênese/genética , Condrogênese/fisiologia , Técnicas de Cocultura , Matriz Extracelular , Fibroínas/química , Humanos , Osteoblastos/fisiologia , Osteogênese/fisiologia , Engenharia Tecidual/métodos
11.
Carbohydr Polym ; 206: 428-434, 2019 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-30553342

RESUMO

Herein, multilayer polysaccharide films were proposed and characterized as biomaterials for the local and controlled release of an antitumoral drug. To that aim, multilayer films of alginate (Alg) and chitosan (Chi) were built up through spray assisted layer-by-layer (LbL) technique employing an automatic equipment. A specific drug against breast cancer, tamoxifen (TMX), was incorporated in different intermediate positions of the multilayer Alg/Chi films. Our findings highlight that Alg/Chi multilayer films can be employed for sustained and local TMX delivery and their therapeutic effect can be modulated and optimized by the number of bilayers deposited over the loaded tamoxifen, the quantity of tamoxifen loaded in several intermediate positions and the total area of the film.


Assuntos
Alginatos/química , Antineoplásicos/farmacologia , Quitosana/química , Preparações de Ação Retardada/química , Tamoxifeno/farmacologia , Alginatos/toxicidade , Antineoplásicos/química , Linhagem Celular Tumoral , Quitosana/toxicidade , Preparações de Ação Retardada/toxicidade , Difusão , Liberação Controlada de Fármacos , Humanos , Tamoxifeno/química , Molhabilidade
12.
PLoS One ; 13(11): e0206515, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30388135

RESUMO

INTRODUCTION: Lightweight (LW) polypropylene (PP) meshes better adapt to host tissue, causing less fibrosis and inflammatory responses than high-density meshes. Mesh fixation using tissue adhesives (TA) that replace conventional sutures may improve the process of hernia repair and tissue trauma. This preclinical study compares the behavior of different cyanoacrylate-based adhesives in the fixation of LW-PP meshes for hernia repair. METHODS: Partial abdominal wall defects were repaired using LW-PP Optilene meshes in New Zealand rabbits. The following groups were established according to the mesh fixation method: Suture (control), Glubran 2 (n-butyl), Ifabond (n-hexyl), SafetySeal (n-butyl) and Evobond (n-octyl). At 14, 90 and 180 days after surgery, the recovered implants were examined to assess the host tissue integration, the macrophage response and the biomechanical strength. RESULTS: All the groups showed optimal host tissue incorporation regardless of the fixation procedure. Significantly increased levels of collagen 1 and collagen 3 gene expression (p<0.001) were observed at 14 days compared to the medium- and long-term durations, where the Suture and Glubran groups showed the highest expression of collagen 1. All the adhesives increased the macrophage reaction (p<0.001) compared to sutures at all implant times. Maximal macrophage response was observed in the short-term Glubran group (p<0.01) compared to the rest of the groups. Although SafetySeal and Evobond did not reach the biomechanical resistance of sutures at 14 days, all the adhesives did reach this level in the medium- to long-term periods, providing significantly higher resistance (p<0.05). CONCLUSIONS: All the cyanoacrylates, despite inducing a significantly increased macrophage response versus sutures, showed optimal host tissue integration and long-term mechanical behavior; thus, they might be good choices for LW-PP mesh hernia repairs.


Assuntos
Cianoacrilatos , Herniorrafia , Polipropilenos , Telas Cirúrgicas , Adesivos , Animais , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Reação a Corpo Estranho/patologia , Macrófagos , Teste de Materiais , Coelhos , Suturas
13.
Biomolecules ; 8(3)2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30235821

RESUMO

The aim of this work is to study, in an in vitro head and neck squamous cell carcinomas model the anti-angiogenic and anti-migratory properties of self-assembled polymeric nanoparticles (NPs) with demonstrated selective anticancer activity. The NPs are based on α-tocopheryl succinate (α-TOS) encapsulated in the hydrophobic core of the NPs. We analyzed the effect of the newly synthetized α-TOS-loaded NPs in proliferating endothelial cells and hypopharynx carcinoma squamous cells and measured markers of angiogenesis, apoptosis and reactive oxygen species (ROS). α-TOS-loaded NPs suppressed angiogenesis by inducing accumulation of ROS and inducing apoptosis of proliferating endothelial cells. These NPs also decrease the number and quality of capillary-like tubes in an in vitro three-dimensional (3D) experiment, decrease the production of the pro-angiogenic vascular endothelial growth factor and down-regulate the expression of its receptor. The anti-migratory efficacy of α-TOS is corroborated in hypopharynx carcinoma cells by decreasing the secretion of matrix metalloproteases 2 and 9 (MMP-2 and MMP-9) and inhibiting cell migration. These results confirm that α-TOS-based NPs not only present anticancer properties, but also antiangiogenic properties, therefore making them promising candidates for multi-active combinatorial anticancer therapy.


Assuntos
Portadores de Fármacos/química , Nanopartículas/química , Polímeros/química , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , alfa-Tocoferol/química , alfa-Tocoferol/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Liberação Controlada de Fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Metaloproteinases da Matriz/metabolismo , Invasividade Neoplásica , Neovascularização Patológica/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , alfa-Tocoferol/uso terapêutico
14.
Polymers (Basel) ; 10(7)2018 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-30960693

RESUMO

The effective treatment of chronic wounds constitutes one of the most common worldwide healthcare problem due to the presence of high levels of proteases, free radicals and exudates in the wound, which constantly activate the inflammatory system, avoiding tissue regeneration. In this study, we describe a multifunctional bioactive and resorbable membrane with in-built antioxidant agent catechol for the continuous quenching of free radicals as well as to control inflammatory response, helping to promote the wound-healing process. This natural polyphenol (catechol) is the key molecule responsible for the mechanism of adhesion of mussels providing also the functionalized polymer with bioadhesion in the moist environment of the human body. To reach that goal, synthesized statistical copolymers of N-vinylcaprolactam (V) and 2-hydroxyethyl methacrylate (H) have been conjugated with catechol bearing hydrocaffeic acid (HCA) molecules with high yields. The system has demonstrated good biocompatibility, a sustained antioxidant response, an anti-inflammatory effect, an ultraviolet (UV) screen, and bioadhesion to porcine skin, all of these been key features in the wound-healing process. Therefore, these novel mussel-inspired materials have an enormous potential for application and can act very positively, favoring and promoting the healing effect in chronic wounds.

15.
J Surg Res ; 220: 30-39, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29180195

RESUMO

BACKGROUND: As an alternative to sutures, meshes used for hernia repair can be fixed using cyanoacrylate-based adhesives. Attempts to improve these adhesives include alkyl-chain lengthening to reduce their toxicity. This preclinical study compares the long-term behavior of cyanoacrylates of different chain lengths already used in hernia repair and new ones for this application. MATERIALS AND METHODS: Partial abdominal wall defects were repaired using a Surgipro mesh in 18 New Zealand White rabbits, and groups were established according to the mesh fixation method: sutures (control), Glubran 2 (n-butyl), Ifabond (n-hexyl), and the new adhesives SafetySeal (n-butyl), and Evobond (n-octyl). Six months after surgery, recovered implants were examined to assess adhesive degradation, host tissue reaction, and biomechanical strength. RESULTS: All the cyanoacrylate groups showed good host tissue incorporation in the meshes. Macrophage responses to Glubran and Ifabond were quantitatively greater compared with sutures. Cell damage caused by the adhesives was similar, and only Glubran induced significantly more damage than sutures. Significantly lower collagen 1/3 messenger RNA expression was induced by Ifabond than the remaining fixation materials. No differences were observed in collagen expression except slightly reduced collagen I deposition in Glubran/Ifabond and collagen III deposition in the suture group. Mechanical strengths failed to vary between the suture and cyanoacrylate groups. CONCLUSIONS: All cyanoacrylates showed good long-term behavior and tolerance irrespective of their long or intermediate chain length. Cyanoacrylate residues persisted at 6 mo, indicating their incomplete degradation. Biomechanical strengths were similar both for the adhesives and sutures.


Assuntos
Cianoacrilatos/uso terapêutico , Hérnia Abdominal/cirurgia , Herniorrafia/instrumentação , Suturas , Animais , Fenômenos Biomecânicos , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Cianoacrilatos/efeitos adversos , Cianoacrilatos/química , Modelos Animais de Doenças , Macrófagos/imunologia , Masculino , Coelhos , Telas Cirúrgicas
17.
J Mater Sci Mater Med ; 28(10): 152, 2017 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-28861765

RESUMO

Active targeting not only of a specific cell but also a specific organelle maximizes the therapeutic activity minimizing adverse side effects in healthy tissues. The present work describes the synthesis, characterization, and in vitro biological activity of active targeting nanoparticles (NP) for cancer therapy based on α-tocopheryl succinate (α-TOS), a well-known mitocan, that selectively induces apoptosis of cancer cells and proliferalting endothelial cells. Human epidermal growth factor receptor 2 (HER2) targeting peptide LTVSPWY (PEP) and triphenylphosphonium lipophilic cation (TPP) were conjugated to a previously optimized RAFT block copolymer that formed self-assembled NP of appropriate size for this application and low polydispersity by self-organized precipitation method. PEP and TPP were included in order to target not only HER2 positive cancer cells, but also the mitochondria of these cancer cells, respectively. The in vitro experiments demonstrated the faster incorporation of the active-targeting NP and the higher accumulation of TPP-bearing NP in the mitochondria of MDA-MB-453 HER2 positive cancer cells compared to non-decorated NP. Moreover, the encapsulation of additional α-TOS in the hydrophobic core of the NP was achieved with high efficiencies. The loaded NP presented higher cytotoxicity than unloaded NP but preserved their selectivity against cancer cells in a range of tested concentrations.


Assuntos
Nanopartículas/química , Oligopeptídeos/química , alfa-Tocoferol/química , Carcinoma , Linhagem Celular Tumoral , Sobrevivência Celular , Corantes Fluorescentes , Humanos , Mitocôndrias/metabolismo , Estrutura Molecular , Oligopeptídeos/farmacologia , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo
18.
Acta Biomater ; 57: 70-84, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28511874

RESUMO

The aim of this work was the generation of a multifunctional nanopolymeric system that incorporates IR-780 dye, a near-infrared (NIR) imaging probe that exhibits photothermal and photodynamic properties; and a derivate of α-tocopheryl succinate (α-TOS), a mitochondria-targeted anticancer compound. IR-780 was conjugated to the hydrophilic segment of copolymer PEG-b-polyMTOS, based on poly(ethylene glycol) (PEG) and a methacrylic derivative of α-tocopheryl succinate (MTOS), to generate IR-NP, self-assembled nanoparticles (NPs) in aqueous media which exhibit a hydrophilic shell and a hydrophobic core. During assembly, the hydrophobic core of IR-NP could encapsulate additional IR-780 to generate derived subspecies carrying different amount of probe (IR-NP-eIR). Evaluation of photo-inducible properties of IR-NP and IR-NP-eIR were thoroughly assessed in vitro. Developed nanotheranostic particles showed distinct fluorescence and photothermal behavior after excitation by a laser light emitting at 808nm. Treatment of MDA-MB-453 cells with IR-NP or IR-NP-eIR resulted in an efficient internalization of the IR-780 dye, while subsequent NIR-laser irradiation led to a severe decrease in cell viability. Photocytoxicity conducted by IR-NP, which could not be attributed to the generation of lethal hyperthermia, responded to an increase in the levels of intracellular reactive oxygen species (ROS). Therefore, the fluorescence imaging and inducible phototoxicity capabilities of NPs derived from IR-780-PEG-b-polyMTOS copolymer confer high value to these nanotheranostics tools in clinical cancer research. STATEMENT OF SIGNIFICANCE: Multifunctional polymeric nanoparticles (NPs) that combine imaging and therapeutic properties are highly valuable in cancer treatment. In this paper we describe the development of NPs that are fluorescent in the near-infrared (NIR). This is important for their visualization in living tissues that present low absorption and low autofluorescence in this wavelength region (between 700 and 1000nm). Moreover, NPs present photothermal and photodynamic properties when NIR irradiated: the NPs produce an efficient increment of temperature and increase the intracellular reactive oxygen species (ROS) when laser irradiated at 808nm. These tuneable photoinduced properties make the NPs highly cytotoxic after NIR irradiation and provide a new tool for highly precise cancer treatment.


Assuntos
Neoplasias da Mama/terapia , Hipertermia Induzida/métodos , Indóis , Nanopartículas , Fotoquimioterapia/métodos , alfa-Tocoferol , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Indóis/química , Indóis/farmacologia , Nanopartículas/química , Nanopartículas/uso terapêutico , alfa-Tocoferol/análogos & derivados , alfa-Tocoferol/química , alfa-Tocoferol/farmacologia
19.
Acta Biomater ; 53: 199-210, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28213099

RESUMO

The aim of this work is the development of highly protective agents to be administered locally within the middle ear to avoid cisplatin-induced ototoxicity, which affects to 100% of the clinical patients at ultra-high concentrations (16mg/kg). The protective agents are based on polymeric nanoparticles loaded with dexamethasone or α-tocopheryl succinate as anti-inflammarory and anti-apoptotic molecules. Dexamethasone and α-tocopheryl succinate are poorly soluble in water and present severe side effects when systemic administered during long periods of time. Their incorporation in the hydrophobic core of nanoparticles with the appropriate hydrodynamic properties provides the desired effects in vitro (lower cisplatin-induced toxicity, decreasing of caspase 3/7 activity, and lower IL-1ß release) and in vivo (reducing the hearing loss at the local level). The local administration of the nanoparticles by bullostomy provides an adequate dose of drug without systemic interference with the chemotherapeutic effect of cisplatin. STATEMENT OF SIGNIFICANCE: 100% of the cancer patients receiving ultra-high doses of CDDP (16mg/kg) suffer severe hearing loss, being a limiting factor in antineoplastic treatments. In this paper we describe the application of polymeric nanoparticles loaded with dexamethasone or α-tocopheryl succinate to palliate the cisplatin ototoxicity derived from chemotherapy treatment. These new nanoparticles, that encapsulate, transport, and deliver dexamethasone or α-tocopheryl succinate in the middle ear, are able to partially prevent ototoxicity derived from high doses of CDDP. This is an interdisciplinary study in which in vitro and in vivo experiments are described and extensively discussed. The importance of the results opens an excellent opportunity to the translation to the clinic.


Assuntos
Cisplatino/antagonistas & inibidores , Cisplatino/toxicidade , Dexametasona/administração & dosagem , Células Ciliadas Auditivas/efeitos dos fármacos , alfa-Tocoferol/administração & dosagem , Animais , Anti-Inflamatórios/administração & dosagem , Apoptose/efeitos dos fármacos , Materiais Biocompatíveis/administração & dosagem , Materiais Biocompatíveis/química , Caspases/metabolismo , Linhagem Celular , Dexametasona/farmacocinética , Sistemas de Liberação de Medicamentos , Potenciais Evocados Auditivos/efeitos dos fármacos , Células Ciliadas Auditivas/patologia , Células Ciliadas Auditivas/fisiologia , Humanos , Interleucina-1beta/metabolismo , Teste de Materiais , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/química , Ratos , Ratos Wistar , alfa-Tocoferol/farmacocinética
20.
J Biomed Mater Res B Appl Biomater ; 105(2): 312-319, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-26505126

RESUMO

AIMS: This study examines the intraperitoneal behavior of two cyanoacrylate tissue adhesives: Ifabond® and a new, non-marketed octyl cyanoacrylate adhesive (OCA) used for the intraperitoneal fixation of a laminar expanded polytetrafluoroethylene (ePTFE) mesh. MATERIAL AND METHODS: In 36 New Zealand White rabbits, 3 × 3 cm (n = 24) or 1.5 × 3 cm (n = 12) fragments of ePTFE mesh (Preclude® , Gore, Flagstaff, USA) were fixed to the parietal peritoneum using OCA or Ifabond® . Peritoneal fluid was obtained at the time of implant and at 2 weeks postimplant for determination of the cytokines interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α). At 14 or 90 days postsurgery, the animals were euthanized and the meshes excised to assess host tissue incorporation, the macrophage response, apoptosis and fixation strength (T-peel tensiometry). RESULTS: Peritoneal fluid IL-6 and TNF-α concentrations were similar in the OCA and Ifabond® groups. Both adhesives gave rise to adequate mesothelialization of the laminar ePTFE. Macrophage counts were similar for the two study groups, but a significantly increase in macrophage response was observed from 14 to 90 days for Ifabond® . At 90 days postimplant, apoptotic cell counts was lower for the implants fixed with OCA and a fixation strength was significantly lower for OCA. CONCLUSIONS: Despite similar cytokine levels at 2 weeks and similar host tissue incorporation observed for the meshes fixed with the two adhesives, the use of Ifabond® gave rise to a greater apoptosis rate, although this adhesive provided a stronger fixation bond. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 312-319, 2017.


Assuntos
Cianoacrilatos , Interleucina-6/metabolismo , Macrófagos/metabolismo , Teste de Materiais , Telas Cirúrgicas , Fator de Necrose Tumoral alfa/metabolismo , Animais , Cianoacrilatos/efeitos adversos , Cianoacrilatos/química , Cianoacrilatos/farmacologia , Polímeros de Fluorcarboneto/efeitos adversos , Polímeros de Fluorcarboneto/química , Polímeros de Fluorcarboneto/farmacologia , Macrófagos/patologia , Coelhos , Adesivos Teciduais/efeitos adversos , Adesivos Teciduais/química , Adesivos Teciduais/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA