Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
ACS Appl Bio Mater ; 7(6): 4175-4192, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38830774

RESUMEN

Nerve growth factor (NGF) plays a crucial role in cellular growth and neurodifferentiation. To achieve significant neuronal regeneration and repair using in vitro NGF delivery, spatiotemporal control that follows the natural neuronal processes must be developed. Notably, a challenge hindering this is the uncontrolled burst release from the growth factor delivery systems. The rapid depletion of NGF reduces treatment efficacy, leading to poor cellular response. To address this, we developed a highly controllable system using graphene oxygen (GO) and GelMA hydrogels modulated by electrical stimulation. Our system showed superior control over the release kinetics, reducing the burst up 30-fold. We demonstrate that the system is also able to sequester and retain NGF up to 10-times more efficiently than GelMA hydrogels alone. Our controlled release system enabled neurodifferentiation, as revealed by gene expression and immunostaining analysis. The increased retention and reduced burst release from our system show a promising pathway for nerve tissue engineering research toward effective regeneration.


Asunto(s)
Materiales Biocompatibles , Estimulación Eléctrica , Grafito , Hidrogeles , Factor de Crecimiento Nervioso , Regeneración Nerviosa , Hidrogeles/química , Hidrogeles/farmacología , Grafito/química , Grafito/farmacología , Regeneración Nerviosa/efectos de los fármacos , Factor de Crecimiento Nervioso/farmacología , Factor de Crecimiento Nervioso/metabolismo , Factor de Crecimiento Nervioso/química , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Animales , Tamaño de la Partícula , Ensayo de Materiales , Ratas , Células PC12 , Ingeniería de Tejidos
2.
ACS Biomater Sci Eng ; 10(6): 3775-3791, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38722625

RESUMEN

This study investigates the electrochemical behavior of GelMA-based hydrogels and their interactions with PC12 neural cells under electrical stimulation in the presence of conducting substrates. Focusing on indium tin oxide (ITO), platinum, and gold mylar substrates supporting conductive scaffolds composed of hydrogel, graphene oxide, and gold nanorods, we explored how the substrate materials affect scaffold conductivity and cell viability. We examined the impact of an optimized electrical stimulation protocol on the PC12 cell viability. According to our findings, substrate selection significantly influences conductive hydrogel behavior, affecting cell viability and proliferation as a result. In particular, the ITO substrates were found to provide the best support for cell viability with an average of at least three times higher metabolic activity compared to platinum and gold mylar substrates over a 7 day stimulation period. The study offers new insights into substrate selection as a platform for neural cell stimulation and underscores the critical role of substrate materials in optimizing the efficacy of neural interfaces for biomedical applications. In addition to extending existing work, this study provides a robust platform for future explorations aimed at tailoring the full potential of tissue-engineered neural interfaces.


Asunto(s)
Supervivencia Celular , Hidrogeles , Neuronas , Compuestos de Estaño , Ingeniería de Tejidos , Andamios del Tejido , Animales , Ingeniería de Tejidos/métodos , Células PC12 , Ratas , Compuestos de Estaño/química , Compuestos de Estaño/farmacología , Hidrogeles/química , Andamios del Tejido/química , Neuronas/fisiología , Neuronas/citología , Oro/química , Oro/farmacología , Grafito/química , Grafito/farmacología , Platino (Metal)/química , Estimulación Eléctrica , Nanotubos/química , Proliferación Celular
3.
Cancers (Basel) ; 15(21)2023 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-37958307

RESUMEN

Myxofibrosarcoma (MFS) is a malignant soft tissue sarcoma (STS) that originates in the body's connective tissues. It is characterized by the presence of myxoid (gel-like) and fibrous components and typically affects patients after the fifth decade of life. Considering the ongoing trend of increasing lifespans across many nations, MFS is likely to become the most common musculoskeletal sarcoma in the future. Although MFS patients have a lower risk of developing distant metastases compared with other STS cases, MFS is characterized by a high frequency of local recurrence. Notably, in 40-60% of the patients where the tumor recurs, it does so multiple times. Consequently, patients may undergo multiple local surgeries, removing the risk of potential amputation. Furthermore, because the tumor relapses generally have a higher grade, they exhibit a decreased response to radio and chemotherapy and an increased tendency to form metastases. Thus, a better understanding of MFS is required, and improved therapeutic options must be developed. Historically, preclinical models for other types of tumors have been instrumental in obtaining a better understanding of tumor development and in testing new therapeutic approaches. However, few MFS models are currently available. In this review, we will describe the MFS models available and will provide insights into the advantages and constraints of each model.

4.
J Mater Chem B ; 11(3): 581-593, 2023 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-36533419

RESUMEN

The development of electroactive cell-laden hydrogels (bioscaffolds) has gained interest in neural tissue engineering research due to their inherent electrical properties that can induce the regulation of cell behaviour. Hydrogels combined with electrically conducting materials can respond to external applied electric fields, where these stimuli can promote electro-responsive cell growth and proliferation. A successful neural interface for electrical stimulation should present the desired stable electrical properties, such as high conductivity, low impedance, increased charge storage capacity and similar mechanical properties related to a target neural tissue. We report how different electrical stimulation protocols can impact neuronal cells' survival and proliferation when using cell-laden GelMA/GO hydrogels. The rat pheochromocytoma cell line, PC12s encapsulated into hydrogels showed an increased proliferation behaviour with increasing current amplitudes applied. Furthermore, the presence of GO in GelMA hydrogels enhanced the metabolic activity and DNA content of PC12s compared with GelMA alone. Similarly, hydrogels provided survival of encapsulated cells at higher current amplitudes when compared to cells seeded onto ITO flat surfaces, which expressed significant cell death at a current amplitude of 2.50 mA. Our findings provide new rational choices for electroactive hydrogels and electrical stimulation with broad potential applications in neural tissue engineering research.


Asunto(s)
Hidrogeles , Andamios del Tejido , Ratas , Animales , Hidrogeles/farmacología , Supervivencia Celular , Estimulación Eléctrica , Proliferación Celular
5.
Target Oncol ; 17(3): 223-252, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35446005

RESUMEN

Sporadic desmoid-type fibromatosis is a rare, fibroblastic soft-tissue neoplasm with local aggressiveness but no metastatic potential. Aberrant Wnt/ß-catenin signalling has been extensively linked to desmoid pathogenesis, although little is known about other molecular drivers and no established treatment approach exists. We aimed to summarise the current literature regarding the molecular pathogenesis of sporadic desmoid-type fibromatosis and to discuss the effects of both current and emerging novel therapies targeting these mechanisms. A literature search was conducted of MEDLINE® ALL and EMBASE databases for published studies (2000-August 2021) using keywords related to 'fibromatosis aggressive', 'immunohistochemistry', 'polymerase chain reaction' and 'mutation'. Articles were included if they examined the role of proteins in sporadic or extra-abdominal human desmoid-type fibromatosis pathogenesis. Searching identified 1684 articles. Following duplicate removal and eligibility screening, 36 were identified. After a full-text screen, 22 were included in the final review. At least 47% of desmoid-type fibromatosis cases displayed aberrant ß-catenin immunoreactivity amongst ten studies. Cyclin D1 overexpression occurred in at least 40% of cases across five studies. Six studies reported oestrogen receptor-ß expression with a range of 7.4-90%. Three studies implicated matrix metalloproteinases, with one study demonstrating vascular endothelial growth factor overexpression. One study explored the positive relationship between cyclooxygenase-2 and platelet-derived growth factor receptor-ß. Aberrant Wnt/ß-catenin signalling is a well-established pathogenic driver that may be targeted via downstream modulation. Growth factor signalling is best appreciated through the clinical trial effects of multi-targeted tyrosine kinase inhibitors, whilst oestrogen receptor expression data may only offer a superficial insight into oestrogen signalling. Finally, the tumour microenvironment presents multiple potential novel therapeutic targets.


Sporadic desmoid tumours are rare soft-tissue neoplasms that arise from connective tissues in the chest wall, head, neck and limbs. Whilst lacking metastatic potential, uncertainty surrounding their locally aggressive growth and unpredictable recurrence complicates treatment approaches. At the molecular level, alterations in the Wnt/ß-catenin signalling pathway, a fundamental coordinator of cell growth and development, have been strongly linked to desmoid tumour development. Beyond this, however, little is known about other molecular drivers. In the case of progressive or life-threatening disease, complex treatment decisions are made regarding the use of surgery, radiotherapy or systemic treatment modalities. Of the targeted systemic therapies, a lack of comparative clinical studies further complicates medical treatment decision making as no definitive treatment approach exists. Therefore, this review aimed to summarise the literature regarding the molecular drivers of desmoid tumour pathogenesis and to discuss the current and emerging novel therapies targeting such mechanisms. Utilising findings from human desmoid tissue samples, we present the rationale for targeting downstream mediators of the central Wnt/ß-catenin pathway and outline potential treatment targets in the tumour microenvironment. We also highlight the knowledge gained from clinical drug trials targeting desmoid growth factor signalling and present the potentially superficial insight provided by oestrogen receptor expression profiles on the role of oestrogen signalling in desmoid pathogenesis. In doing so, this work may assist in the eventual development of an evidence-based treatment approach for sporadic desmoid tumours.


Asunto(s)
Fibromatosis Agresiva , beta Catenina , Fibromatosis Agresiva/tratamiento farmacológico , Fibromatosis Agresiva/genética , Fibromatosis Agresiva/metabolismo , Humanos , Receptores de Estrógenos , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular , Vía de Señalización Wnt , beta Catenina/metabolismo
6.
Pharmaceutics ; 14(3)2022 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-35336051

RESUMEN

Osteosarcoma treatment is moving towards more effective combination therapies. Nevertheless, these approaches present distinctive challenges that can complicate the clinical translation, such as increased toxicity and multi-drug resistance. Drug co-encapsulation within a nanoparticle formulation can overcome these challenges and improve the therapeutic index. We previously synthetized keratin nanoparticles functionalized with Chlorin-e6 (Ce6) and paclitaxel (PTX) to combine photo (PDT) and chemotherapy (PTX) regimens, and the inhibition of osteosarcoma cells growth in vitro was demonstrated. In the current study, we generated an orthotopic osteosarcoma murine model for the preclinical evaluation of our combination therapy. To achieve maximum reproducibility, we systematically established key parameters, such as the number of cells to generate the tumor, the nanoparticles dose, the design of the light-delivery device, the treatment schedule, and the irradiation settings. A 60% engrafting rate was obtained using 10 million OS cells inoculated intratibial, with the tumor model recapitulating the histological hallmarks of the human counterpart. By scheduling the treatment as two cycles of injections, a 32% tumor reduction was obtained with PTX mono-therapy and a 78% reduction with the combined PTX-PDT therapy. Our findings provide the in vivo proof of concept for the subsequent clinical development of a combination therapy to fight osteosarcoma.

7.
Int J Mol Sci ; 22(22)2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-34830302

RESUMEN

Osteochondral (OC) defects are debilitating joint injuries characterized by the loss of full thickness articular cartilage along with the underlying calcified cartilage through to the subchondral bone. While current surgical treatments can provide some relief from pain, none can fully repair all the components of the OC unit and restore its native function. Engineering OC tissue is challenging due to the presence of the three distinct tissue regions. Recent advances in additive manufacturing provide unprecedented control over the internal microstructure of bioscaffolds, the patterning of growth factors and the encapsulation of potentially regenerative cells. These developments are ushering in a new paradigm of 'multiphasic' scaffold designs in which the optimal micro-environment for each tissue region is individually crafted. Although the adoption of these techniques provides new opportunities in OC research, it also introduces challenges, such as creating tissue interfaces, integrating multiple fabrication techniques and co-culturing different cells within the same construct. This review captures the considerations and capabilities in developing 3D printed OC scaffolds, including materials, fabrication techniques, mechanical function, biological components and design.


Asunto(s)
Enfermedades de los Cartílagos/cirugía , Trasplante de Células Madre Mesenquimatosas/métodos , Impresión Tridimensional , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Implantes Absorbibles , Animales , Materiales Biocompatibles , Huesos , Cartílago Articular , Humanos , Trasplante de Tejidos/métodos
8.
Tissue Eng Part B Rev ; 27(5): 514-529, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33138724

RESUMEN

Osteosarcoma (OS) is a highly aggressive primary bone tumor. The mainstay for its treatment is multiagent chemotherapy and surgical resection, with a 50-70% 5-year survival rate. Despite the huge effort made by clinicians and researchers in the past 30 years, limited progress has been made to improve patient outcomes. As novel therapeutic approaches for OS become available, such as monoclonal antibodies, small molecules, and immunotherapies, the need for OS preclinical model development becomes equally pressing. Three-dimensional (3D) OS models represent an alternative system to study this tumor: In contrast to two-dimensional monolayers, 3D matrices can recapitulate key elements of the tumor microenvironment (TME), such as the cellular interaction with the bone mineralized matrix. The advancement of tissue engineering and biofabrication techniques enables the incorporation of specific TME aspects into 3D models, to investigate the contribution of individual components to tumor progression and enhance understanding of basic OS biology. The use of biomaterials that mimic the extracellular matrix could also facilitate the testing of drugs targeting the TME itself, allowing a larger range of therapeutics to be tested, while averting the ethical implications and high cost associated with in vivo preclinical models. This review aims at serving as a practical guide by delineating the OS TME ("what it is like") and, in turn, propose various biofabrication strategies to create a 3D model ("how to recreate it"), to improve the in vitro representation of the OS tumor and ultimately generate more accurate drug response profiles.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Neoplasias Óseas/terapia , Comunicación Celular , Humanos , Osteosarcoma/terapia , Ingeniería de Tejidos , Microambiente Tumoral
9.
Cancers (Basel) ; 12(8)2020 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-32824479

RESUMEN

Bone is one of the most common sites for cancer metastasis. Bone tissue is composed by different kinds of cells that coexist in a coordinated balance. Due to the complexity of bone, it is impossible to capture the intricate interactions between cells under either physiological or pathological conditions. Hence, a variety of in vivo and in vitro approaches have been developed. Various models of tumor-bone diseases are routinely used to provide valuable information on the relationship between metastatic cancer cells and the bone tissue. Ideally, when modeling the metastasis of human cancers to bone, models would replicate the intra-tumor heterogeneity, as well as the genetic and phenotypic changes that occur with human cancers; such models would be scalable and reproducible to allow high-throughput investigation. Despite the continuous progress, there is still a lack of solid, amenable, and affordable models that are able to fully recapitulate the biological processes happening in vivo, permitting a correct interpretation of results. In the last decades, researchers have demonstrated that three-dimensional (3D) methods could be an innovative approach that lies between bi-dimensional (2D) models and animal models. Scientific evidence supports that the tumor microenvironment can be better reproduced in a 3D system than a 2D cell culture, and the 3D systems can be scaled up for drug screening in the same way as the 2D systems thanks to the current technologies developed. However, 3D models cannot completely recapitulate the inter- and intra-tumor heterogeneity found in patients. In contrast, ex vivo cultures of fragments of bone preserve key cell-cell and cell-matrix interactions and allow the study of bone cells in their natural 3D environment. Moreover, ex vivo bone organ cultures could be a better model to resemble the human pathogenic metastasis condition and useful tools to predict in vivo response to therapies. The aim of our review is to provide an overview of the current trends in bone metastasis modeling. By showing the existing in vitro and ex vivo systems, we aspire to contribute to broaden the knowledge on bone metastasis models and make these tools more appealing for further translational studies.

10.
Methods Mol Biol ; 2140: 145-157, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32207110

RESUMEN

Three-dimensional (3D) bioprinting is driving major innovations in the area of cartilage tissue engineering. As an alternative to computer-aided 3D printing, in situ additive manufacturing has the advantage of matching the geometry of the defect to be repaired without specific preliminary image analysis, shaping the bioscaffold within the defect, and achieving the best possible contact between the bioscaffold and the host tissue. Here, we describe an in situ approach that allows 3D bioprinting of human adipose-derived stem cells (hADSCs) laden in 10%GelMa/2%HAMa (GelMa/HAMa) hydrogel. We use coaxial extrusion to obtain a core/shell bioscaffold with high cell viability, as well as adequate mechanical properties for articular cartilage regeneration and repair.


Asunto(s)
Materiales Biocompatibles , Bioimpresión/métodos , Cartílago Articular/cirugía , Regeneración Tisular Dirigida/métodos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Impresión Tridimensional , Supervivencia Celular , Humanos , Hidrogeles/efectos de la radiación , Células Madre Mesenquimatosas/citología , Metacrilatos , Fotoquímica , Ingeniería de Tejidos , Andamios del Tejido
11.
J Exp Clin Cancer Res ; 39(1): 40, 2020 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-32087737

RESUMEN

BACKGROUND: Osteosarcoma (OS) is an aggressive malignant neoplasm that still suffers from poor prognosis in the case of distal metastases or occurrence of multi-drug resistance. It is therefore crucial to find novel therapeutic options able to go beyond these limitations and improve patients' survival. The objective of this study is to exploit the intrinsic properties of mesenchymal stromal cells (MSCs) to migrate and infiltrate the tumor stroma to specifically deliver therapeutic agents directly to cancer cells. In particular, we aimed to test the efficacy of the photoactivation of MSCs loaded with nanoparticles in vitro and in a murine in vivo ectopic osteosarcoma model. METHODS: AlPcS4@FNPs were produced by adding tetra-sulfonated aluminum phthalocyanine (AlPcS4) to an aqueous solution of positively charged poly-methyl methacrylate core-shell fluorescent nanoparticles (FNPs). The photodynamic therapy (PDT) effect is achieved by activation of the photosensitizer AlPcS4 in the near-infrared light with an LED source. Human MSCs were isolated from the bone marrow of five donors to account for inter-patients variability and used in this study after being evaluated for their clonogenicity, multipotency and immunophenotypic profile. MSC lines were then tested for the ability to internalize and retain the nanoparticles, along with their migratory properties in vitro. Photoactivation effect was evaluated both in a monolayer (2D) co-culture of AlPcS4@FNPs loaded MSCs with human OS cells (SaOS-2) and in tridimensional (3D) multicellular spheroids (AlPcS4@FNPs loaded MSCs with human OS cells, MG-63). Cell death was assessed by AnnexinV/PI and Live&Dead CalceinAM/EthD staining in 2D, while in the 3D co-culture, the cell killing effect was measured through ATP content, CalceinAM/EthD staining and TEM imaging. We also evaluated the effectiveness of AlPcS4@FNPs loaded MSCs as delivery systems and the ability of the photodynamic treatment to kill cancer cells in a subcutaneous mouse model of OS by bioluminescence imaging (BLI) and histology. RESULTS: MSCs internalized AlPcS4@FNPs without losing or altering their motility and viability in vitro. Photoactivation of AlPcS4@FNPs loaded MSCs induced high level of OS cells death in the 2D co-culture. Similarly, in the 3D co-culture (MSCs:OS ratios 1:1 or 1:3), a substantial decrease of both MSCs and OS cells viability was observed. Notably, when increasing the MSCs:OS ratio to 1:7, photoactivation still caused more than 40% cells death. When tested in an in vivo ectopic OS model, AlPcS4@FNPs loaded MSCs were able to decrease OS growth by 68% after two cycles of photoactivation. CONCLUSIONS: Our findings demonstrate that MSCs can deliver functional photosensitizer-decorated nanoparticles in vitro and in vivo and inhibit OS tumor growth. MSCs may be an effective platform for the targeted delivery of therapeutic nanodrugs in a clinical scenario, alone or in combination with other osteosarcoma treatment modalities.


Asunto(s)
Neoplasias Óseas/terapia , Indoles/administración & dosificación , Células Madre Mesenquimatosas/citología , Compuestos Organometálicos/administración & dosificación , Osteosarcoma/terapia , Fármacos Fotosensibilizantes/administración & dosificación , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Humanos , Indoles/farmacología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/química , Ratones , Nanopartículas , Compuestos Organometálicos/farmacología , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Biofabrication ; 11(3): 035003, 2019 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-30818298

RESUMEN

Reliable and scalable sterilisation of hydrogels is critical to the clinical translation of many biofabrication approaches, such as extrusion-based 3D bioprinting of cell-laden bio-inks. However sterilisation methods can be destructive, and may have detrimental effects on the naturally-derived hydrogels that constitute much of the bio-ink palette. Determining effective sterilisation methods requires detailed analysis of the effects of sterilisation on relevant properties such as viscosity, printability and cytocompatibility. Yet there have been no studies specifically exploring the effects of sterilisation on bio-inks to date. In this work, we explored the effects of various sterilisation techniques on four of the most widely used bio-ink components: gelatin, gelatin methacryloyl, hyaluronic acid, and hyaluronic acid methacrylate. Autoclaving was the most destructive sterilisation method, producing large reductions in viscosity and in mechanical properties following crosslinking. Filter sterilisation caused some reduction in rheological properties of GelMA due to removal of higher molecular weight components, but did not affect photocrosslinking. Ethylene oxide (EtO) was the least destructive sterilisation method in terms of rheological properties for all materials, had no detrimental effect on the photocrosslinkable methacrylate/methacrylamide groups, and so was chosen for more detailed examination. In biological analyses, we found that EtO treatment successfully eradicated a bacterial challenge of E. coli, caused no decrease in viability of human mesenchyman stem cells (hMSCs), and had no effect on their rate of proliferation. Finally, we found that EtO-treated hydrogels supported encapsulated hMSCs to differentiate towards the chondrogenic lineage, and to produce new cartilage matrix. Our results bring to light various effects that sterilisation can have on bio-inks, as well as highlighting EtO sterilisation as a method which minimises degradation of properties, while still promoting biological function.


Asunto(s)
Gelatina/farmacología , Ácido Hialurónico/farmacología , Tinta , Metacrilatos/farmacología , Esterilización/métodos , Tejido Adiposo/citología , Animales , Cartílago/efectos de los fármacos , Supervivencia Celular , Reactivos de Enlaces Cruzados/química , Óxido de Etileno/química , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Células Madre Mesenquimatosas/citología , Microtecnología , Reología , Porcinos
13.
Int J Mol Sci ; 19(11)2018 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-30463350

RESUMEN

Osteosarcoma therapy might be moving toward nanotechnology-based drug delivery systems to reduce the cytotoxicity of antineoplastic drugs and improve their pharmacokinetics. In this paper, we present, for the first time, an extensive chemical and in vitro characterization of dual-loaded photo- and chemo-active keratin nanoparticles as a novel drug delivery system to treat osteosarcoma. The nanoparticles are prepared from high molecular weight and hydrosoluble keratin, suitably functionalized with the photosensitizer Chlorin-e6 (Ce6) and then loaded with the chemotherapeutic drug Paclitaxel (PTX). This multi-modal PTX-Ce6@Ker nanoformulation is prepared by both drug-induced aggregation and desolvation methods, and a comprehensive physicochemical characterization is performed. PTX-Ce6@Ker efficacy is tested on osteosarcoma tumor cell lines, including chemo-resistant cells, using 2D and 3D model systems. The single and combined contributions of PTX and Ce6 is evaluated, and results show that PTX retains its activity while being vehiculated through keratin. Moreover, PTX and Ce6 act in an additive manner, demonstrating that the combination of the cytostatic blockage of PTX and the oxidative damage of ROS upon light irradiation have a far superior effect compared to singularly administered PTX or Ce6. Our findings provide the proof of principle for the development of a novel, nanotechnology-based drug delivery system for the treatment of osteosarcoma.


Asunto(s)
Sistemas de Liberación de Medicamentos , Queratinas/química , Nanotecnología , Osteosarcoma/tratamiento farmacológico , Línea Celular Tumoral , Supervivencia Celular , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Nanopartículas/química , Nanopartículas/toxicidad , Nanopartículas/ultraestructura , Osteosarcoma/patología , Paclitaxel/farmacología
14.
Biofabrication ; 10(4): 045006, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30088479

RESUMEN

Cartilage injuries cause pain and loss of function, and if severe may result in osteoarthritis (OA). 3D bioprinting is now a tangible option for the delivery of bioscaffolds capable of regenerating the deficient cartilage tissue. Our team has developed a handheld device, the Biopen, to allow in situ additive manufacturing during surgery. Given its ability to extrude in a core/shell manner, the Biopen can preserve cell viability during the biofabrication process, and it is currently the only biofabrication tool tested as a surgical instrument in a sheep model using homologous stem cells. As a necessary step toward the development of a clinically relevant protocol, we aimed to demonstrate that our handheld extrusion device can successfully be used for the biofabrication of human cartilage. Therefore, this study is a required step for the development of a surgical treatment in human patients. In this work we specifically used human adipose derived mesenchymal stem cells (hADSCs), harvested from the infra-patellar fat pad of donor patients affected by OA, to also prove that they can be utilized as the source of cells for the future clinical application. With the Biopen, we generated bioscaffolds made of hADSCs laden in gelatin methacrylate, hyaluronic acid methacrylate and cultured in the presence of chondrogenic stimuli for eight weeks in vitro. A comprehensive characterisation including gene and protein expression analyses, immunohistology, confocal microscopy, second harmonic generation, light sheet imaging, atomic force mycroscopy and mechanical unconfined compression demonstrated that our strategy resulted in human hyaline-like cartilage formation. Our in situ biofabrication approach represents an innovation with important implications for customizing cartilage repair in patients with cartilage injuries and OA.


Asunto(s)
Cartílago Articular/fisiología , Ingeniería de Tejidos/métodos , Tejido Adiposo/citología , Diferenciación Celular/genética , Condrogénesis/genética , Colágeno Tipo II/metabolismo , Gelatina/química , Regulación de la Expresión Génica , Glicosaminoglicanos/metabolismo , Humanos , Ácido Hialurónico/química , Metacrilatos/química , Células Madre/metabolismo
15.
J Tissue Eng Regen Med ; 12(3): 611-621, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28512850

RESUMEN

Articular cartilage injuries experienced at an early age can lead to the development of osteoarthritis later in life. In situ three-dimensional (3D) printing is an exciting and innovative biofabrication technology that enables the surgeon to deliver tissue-engineering techniques at the time and location of need. We have created a hand-held 3D printing device (biopen) that allows the simultaneous coaxial extrusion of bioscaffold and cultured cells directly into the cartilage defect in vivo in a single-session surgery. This pilot study assessed the ability of the biopen to repair a full-thickness chondral defect and the early outcomes in cartilage regeneration, and compared these results with other treatments in a large animal model. A standardized critical-sized full-thickness chondral defect was created in the weight-bearing surface of the lateral and medial condyles of both femurs of six sheep. Each defect was treated with one of the following treatments: (i) hand-held in situ 3D printed bioscaffold using the biopen (HH group), (ii) preconstructed bench-based printed bioscaffolds (BB group), (iii) microfractures (MF group) or (iv) untreated (control, C group). At 8 weeks after surgery, macroscopic, microscopic and biomechanical tests were performed. Surgical 3D bioprinting was performed in all animals without any intra- or postoperative complication. The HH biopen allowed early cartilage regeneration. The results of this study show that real-time, in vivo bioprinting with cells and scaffold is a feasible means of delivering a regenerative medicine strategy in a large animal model to regenerate articular cartilage.


Asunto(s)
Bioimpresión , Cartílago Articular/fisiología , Impresión Tridimensional , Regeneración/fisiología , Animales , Fenómenos Biomecánicos , Cartílago Articular/cirugía , Masculino , Células Madre Mesenquimatosas/citología , Ovinos , Ingeniería de Tejidos
16.
Sci Rep ; 7(1): 16820, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29203880

RESUMEN

The abnormal wing discs (awd) gene encodes the Drosophila homolog of NME1/NME2 metastasis suppressor genes. Awd acts in multiple tissues where its function is critical in establishing and maintaining epithelial integrity. Here, we analysed awd gene function in Drosophila epithelial cells using transgene-mediated RNA interference and genetic mosaic analysis. We show that awd knockdown in larval wing disc epithelium leads to chromosomal instability (CIN) and induces apoptosis mediated by activation of c-Jun N-terminal kinase. Forced maintenance of Awd depleted cells, by expressing the cell death inhibitor p35, downregulates atypical protein kinase C and DE-Cadherin. Consistent with their loss of cell polarity and enhanced level of matrix metalloproteinase 1, cells delaminate from wing disc epithelium. Furthermore, the DNA content profile of these cells indicates that they are aneuploid. Overall, our data demonstrate a novel function for awd in maintenance of genomic stability. Our results are consistent with other studies reporting that NME1 down-regulation induces CIN in human cell lines and suggest that Drosophila model could be successfully used to study in vivo the impact of NME/Awd - induced genomic instability on tumour development and metastasis formation.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/genética , Inestabilidad Genómica , Nucleósido-Difosfato Quinasa/genética , Aneuploidia , Animales , Cadherinas/metabolismo , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Larva/crecimiento & desarrollo , Larva/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Microscopía Fluorescente , Nucleósido-Difosfato Quinasa/antagonistas & inhibidores , Nucleósido-Difosfato Quinasa/metabolismo , Proteína Quinasa C/metabolismo , Interferencia de ARN , Alas de Animales/metabolismo , Alas de Animales/patología
17.
PLoS One ; 12(8): e0183336, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28817694

RESUMEN

Cell interaction with biomaterials is one of the keystones to developing medical devices for tissue engineering applications. Biomaterials are the scaffolds that give three-dimensional support to the cells, and are vectors that deliver the cells to the injured tissue requiring repair. Features of biomaterials can influence the behaviour of the cells and consequently the efficacy of the tissue-engineered product. The adhesion, distribution and motility of the seeded cells onto the scaffold represent key aspects, and must be evaluated in vitro during the product development, especially when the efficacy of a specific tissue-engineered product depends on viable and functional cell loading. In this work, we propose a non-invasive and non-destructive imaging analysis for investigating motility, viability and distribution of Mesenchymal Stem Cells (MSCs) on silk fibroin-based alginate microcarriers, to test the adhesion capacity of the fibroin coating onto alginate which is known to be unsuitable for cell adhesion. However, in depth characterization of the biomaterial is beyond the scope of this paper. Scaffold-loaded MSCs were stained with Calcein-AM and Ethidium homodimer-1 to detect live and dead cells, respectively, and counterstained with Hoechst to label cell nuclei. Time-lapse Light Sheet Fluorescent Microscopy (LSFM) was then used to produce three-dimensional images of the entire cells-loaded fibroin/alginate microcarriers. In order to quantitatively track the cell motility over time, we also developed an open source user friendly software tool called Fluorescent Cell Tracker in Three-Dimensions (F-Tracker3D). Combining LSFM with F-Tracker3D we were able for the first time to assess the distribution and motility of stem cells in a non-invasive, non-destructive, quantitative, and three-dimensional analysis of the entire surface of the cell-loaded scaffold. We therefore propose this imaging technique as an innovative holistic tool for monitoring cell-biomaterial interactions, and as a tool for the design, fabrication and functionalization of a scaffold as a medical device.


Asunto(s)
Alginatos/química , Fibroínas/química , Microscopía Fluorescente/métodos , Adhesión Celular , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Humanos , Células Madre Mesenquimatosas/citología , Ingeniería de Tejidos/métodos
18.
Sci Rep ; 7(1): 5837, 2017 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-28724980

RESUMEN

Three-dimensional (3D) bioprinting is driving major innovations in the area of cartilage tissue engineering. Extrusion-based 3D bioprinting necessitates a phase change from a liquid bioink to a semi-solid crosslinked network achieved by a photo-initiated free radical polymerization reaction that is known to be cytotoxic. Therefore, the choice of the photocuring conditions has to be carefully addressed to generate a structure stiff enough to withstand the forces phisiologically applied on articular cartilage, while ensuring adequate cell survival for functional chondral repair. We recently developed a handheld 3D printer called "Biopen". To progress towards translating this freeform biofabrication tool into clinical practice, we aimed to define the ideal bioprinting conditions that would deliver a scaffold with high cell viability and structural stiffness relevant for chondral repair. To fulfill those criteria, free radical cytotoxicity was confined by a co-axial Core/Shell separation. This system allowed the generation of Core/Shell GelMa/HAMa bioscaffolds with stiffness of 200KPa, achieved after only 10 seconds of exposure to 700 mW/cm2 of 365 nm UV-A, containing >90% viable stem cells that retained proliferative capacity. Overall, the Core/Shell handheld 3D bioprinting strategy enabled rapid generation of high modulus bioscaffolds with high cell viability, with potential for in situ surgical cartilage engineering.


Asunto(s)
Bioimpresión/métodos , Cartílago Articular/cirugía , Regeneración , Animales , Muerte Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Módulo de Elasticidad , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Luz , Metacrilatos/química , Ovinos , Andamios del Tejido/química
19.
Life Sci ; 173: 28-35, 2017 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-28131762

RESUMEN

AIM: Mesenchymal stem cells (MSC) possess the distinctive feature of homing in on and engrafting into the tumor stroma making their therapeutic applications in cancer treatment very promising. Research into new effectors and external stimuli, which can selectively trigger the release of cytotoxic species from MSC toward the cancer cells, significantly raises their potential. MAIN METHODS: Shock waves (SW) have recently gained recognition for their ability to induce specific biological effects, such as the local generation of cytotoxic reactive oxygen species (ROS) in a non-invasive and tunable manner. We thus investigate whether MSC are able to generate ROS and, in turn, affect cancer cell growth when in co-culture with human glioblastoma (U87) or osteosarcoma (U2OS) cells and exposed to SW. KEY FINDINGS: MSC were found to be the cell line that was most sensitive to SW treatment as shown by SW-induced ROS production and cytotoxicity. Notably, U87 and U2OS cancer cell growth was unaffected by SW exposure. However, significant decreases in cancer cell growth, 1.8 fold for U87 and 2.3 fold for U2OS, were observed 24h after the SW treatment of MSC co-cultures with cancer cells. The ROS production induced in MSC by SW exposure was then responsible for lipid peroxidation and cell death in U87 and U2OS cells co-cultured with MSC. SIGNIFICANCE: This experiment highlights the unique ability of MSC to generate ROS upon SW treatment and induce the cell death of co-cultured cancer cells. SW might therefore be proposed as an innovative tool for MSC-mediated cancer treatment.


Asunto(s)
Neoplasias Óseas , Glioblastoma , Ondas de Choque de Alta Energía , Mecanotransducción Celular , Células Madre Mesenquimatosas/metabolismo , Osteosarcoma , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/terapia , Muerte Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Peroxidación de Lípido , Masculino , Células Madre Mesenquimatosas/patología , Osteosarcoma/metabolismo , Osteosarcoma/patología , Osteosarcoma/terapia , Especies Reactivas de Oxígeno/metabolismo
20.
Nanomedicine ; 12(7): 1885-1897, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27133189

RESUMEN

Conventional photodynamic therapy has shown to be beneficial in the treatment of a variety of tumors. However, one of its major limitations is the inadequate penetration depth of visible light. In order to overcome this constraint, we developed 80nm poly-methylmethacrylate core-shell fluorescent nanoparticles (FNP) loaded with the photosensitizer tetrasulfonated aluminum phthalocyanine (Ptl). To demonstrate the efficacy of our Ptl@FNP we performed in vitro and in vivo studies using a human prostate tumor model. Our data reveal that Ptl@FNP are internalized by tumor cells, favour Ptl intracellular accumulation, and efficiently trigger cell death through the generation of ROS upon irradiation with 680nm light. When directly injected into tumors intramuscularly induced in SCID mice, Ptl@FNP upon irradiation significantly reduce tumor growth with higher efficiency than the bare Ptl. Collectively, these results demonstrate that the newly developed nanoparticles may be utilized as a delivery system for antitumor phototherapy in solid cancers.


Asunto(s)
Indoles/administración & dosificación , Nanopartículas , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Línea Celular Tumoral , Humanos , Isoindoles , Masculino , Ratones , Ratones SCID
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA