Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
2.
PLoS One ; 19(5): e0303106, 2024.
Article in English | MEDLINE | ID: mdl-38691566

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0083734.].

3.
Nanomaterials (Basel) ; 13(18)2023 Sep 08.
Article in English | MEDLINE | ID: mdl-37764548

ABSTRACT

Three-dimensional (3D) printing technology has become a popular tool to produce complex structures. It has great potential in the regenerative medicine field to produce customizable and reproducible scaffolds with high control of dimensions and porosity. This study was focused on the investigation of new biocompatible and biodegradable 3D-printed scaffolds with suitable mechanical properties to assist tendon and ligament regeneration. Polylactic acid (PLA) scaffolds were reinforced with 0.5 wt.% of functionalized graphite nanoplatelets decorated with silver nanoparticles ((f-EG)+Ag). The functionalization of graphene was carried out to strengthen the interface with the polymer. (f-EG)+Ag exhibited antibacterial properties against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli), an important feature for the healing process and prevention of bacterial infections. The scaffolds' structure, biodegradation, and mechanical properties were assessed to confirm their suitability for tendon and ligamentregeneration. All scaffolds exhibited surface nanoroughness created during printing, which was increased by the filler presence. The wet state dynamic mechanical analysis proved that the incorporation of reinforcement led to an increase in the storage modulus, compared with neat PLA. The cytotoxicity assays using L929 fibroblasts showed that the scaffolds were biocompatible. The PLA+[(f-EG)+Ag] scaffolds were also loaded with human tendon-derived cells and showed their capability to maintain the tenogenic commitment with an increase in the gene expression of specific tendon/ligament-related markers. The results demonstrate the potential application of these new 3D-printed nanocomposite scaffolds for tendon and ligament regeneration.

4.
Article in English | MEDLINE | ID: mdl-37527508

ABSTRACT

In recent years, nanotechnology-based microRNA (miR) therapeutic platforms have shown great promise for immunotherapy and tissue regeneration, despite the unmet challenge of achieving efficient and safe delivery of miRs. The transport of miRs offers precision and regulatory value for a myriad of biological processes and pathways, including the control of macrophage (Mφ) functions and, consequently, the inflammatory cascades Mφ are involved in. Thus, enforcement of Mφ can boost the regenerative process and provide new solutions for diverse chronic pathologies. In this study, we sought to develop a magnetically guided transporter to deliver an miR-155 antagonist to M1-primed Mφ. Furthermore, we determined its modulatory effect in reprogramming Mφ from inflammatory to pro-regenerative phenotypes, with the aim of tissue healing and regenerative medicine approaches. This strategy combines contactless and high-precision control of Mφ, anticipating new functional miR carriers for targeted strategies controlled by extracorporeal action. The magnetoplexes SPION@PEI-miR were efficiently delivered into Mφ without compromising cell viability and successfully induced miR-mediated gene silencing by enhancing the expression of anti-inflammatory markers (IL4 and IL10) and the production of M2φ-related markers (CD206 and IL4). Given its multimodal features, SPION@PEI-miR represents a simple, safe, and nonviral theranostic platform that enables imaging, tracking, and miR delivery with modulatory effects on immune cells.

5.
Adv Drug Deliv Rev ; 196: 114815, 2023 05.
Article in English | MEDLINE | ID: mdl-37001644

ABSTRACT

Tendon afflictions constitute a significant share of musculoskeletal diseases and represent a primary cause of incapacity worldwide. Unresolved/chronic inflammatory states have been associated with the onset and progression of tendon disorders, contributing to undesirable immune stimulation and detrimental tissue effects. Thus, targeting persistent inflammatory events could assist important developments to solve pathophysiological processes and innovative therapeutics to address impaired healing and accomplish complete tendon regeneration. This review overviews the impact of inflammation and inflammatory mediators in tendon niches, unveiling the importance of tendon cell populations and their signature features, and the influence of microenvironmental factors on inflamed and injured tendons. The demand for non-invasive instructive strategies to manage persistent inflammatory mediators, guide inflammatory pathways, and modulate cellular responses will also be approached by exploring the role of pulsed electromagnetic field (PEMF). PEMF alone or combined with more sophisticated systems triggered by magnetic fields will be considered in the design of successful therapies to control inflammation in tendinopathic conditions.


Subject(s)
Tendons , Wound Healing , Humans , Electromagnetic Fields , Magnetic Fields , Inflammation/therapy
6.
Int J Mol Sci ; 23(23)2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36499452

ABSTRACT

The persistence of inflammatory mediators in tissue niches significantly impacts regenerative outcomes and contributes to chronic diseases. Interleukin-4 (IL4) boosts pro-healing phenotypes in macrophages (Mφ) and triggers the activation of signal transducer and activator of transcription 6 (STAT6). Since the IL4/STAT6 pathway reduces Mφ responsiveness to inflammation in a targeted and precise manner, IL4 delivery offers personalized possibilities to overcome inflammatory events. Despite its therapeutic potential, the limited success of IL4-targeted delivery is hampered by inefficient vehicles. Magnetically assisted technologies offer precise and tunable nanodevices for the delivery of cytokines by combining contactless modulation, high tissue penetration, imaging features, and low interference with the biological environment. Although superparamagnetic iron oxide nanoparticles (SPION) have shown clinical applicability in imaging, SPION-based approaches have rarely been explored for targeted delivery and cell programming. Herein, we hypothesized that SPION-based carriers assist in efficient IL4 delivery to Mφ, favoring a pro-regenerative phenotype (M2φ). Our results confirmed the efficiency of SPION-IL4 and Mφ responsiveness to SPION-IL4 with evidence of STAT6-mediated polarization. SPION-IL4-treated Mφ showed increased expression of M2φ associated-mediators (IL10, ARG1, CCL2, IL1Ra) when compared to the well-established soluble IL4. The ability of SPION-IL4 to direct Mφ polarization using sophisticated magnetic nanotools is valuable for resolving inflammation and assisting innovative strategies for chronic inflammatory conditions.


Subject(s)
Macrophage Activation , Nanoparticles , Humans , Macrophages/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism
7.
Int J Mol Sci ; 23(19)2022 Oct 04.
Article in English | MEDLINE | ID: mdl-36233094

ABSTRACT

Hybrid nanoarchitectures such as magnetic polymeric micelles (MPMs) are among the most promising nanotechnology-enabled materials for biomedical applications combining the benefits of polymeric micelles and magnetic nanoparticles within a single bioinstructive system. MPMs are formed by the self-assembly of polymer amphiphiles above the critical micelle concentration, generating a colloidal structure with a hydrophobic core and a hydrophilic shell incorporating magnetic particles (MNPs) in one of the segments. MPMs have been investigated most prominently as contrast agents for magnetic resonance imaging (MRI), as heat generators in hyperthermia treatments, and as magnetic-susceptible nanocarriers for the delivery and release of therapeutic agents. The versatility of MPMs constitutes a powerful route to ultrasensitive, precise, and multifunctional diagnostic and therapeutic vehicles for the treatment of a wide range of pathologies. Although MPMs have been significantly explored for MRI and cancer therapy, MPMs are multipurpose functional units, widening their applicability into less expected fields of research such as bioengineering and regenerative medicine. Herein, we aim to review published reports of the last five years about MPMs concerning their structure and fabrication methods as well as their current and foreseen expectations for advanced biomedical applications.


Subject(s)
Hyperthermia, Induced , Micelles , Contrast Media , Drug Delivery Systems/methods , Polymers/chemistry , Precision Medicine
8.
Nanoscale ; 14(17): 6543-6556, 2022 May 05.
Article in English | MEDLINE | ID: mdl-35420605

ABSTRACT

Extracellular vesicles (EVs) have emerged as cell-free nanotherapeutic agents for the potential treatment of multiple diseases and for tissue engineering and regenerative medicine strategies. Nevertheless, the field has typically relied on EVs derived from stem cells, the production of which in high quantities and high reproducibility is still under debate. Platelet-derived EVs were produced by a freeze-thaw method of platelet concentrates, a highly available clinical waste material. The aim of this study was to produce and thoroughly characterize platelet-derived EVs and understand their effects in adipose-tissue derived stem cells (hASCs), endothelial cells (HUVECs) and macrophages. Two different EV populations were obtained after differential centrifugation, namely small EVs (sEVs) and medium EVs (mEVs), which showed different size distributions and unique proteomic signatures. EV interaction with hASCs resulted in the modulation of the gene expression of markers related to their commitment toward different lineages. Moreover, mEVs showed higher angiogenic potential than sEVs, in a tube formation assay with HUVECs. Also, the EVs were able to modulate macrophage polarization. Altogether, these results suggest that platelet-derived EVs are promising candidates to be used as biochemical signals or therapeutic tools in tissue engineering and regenerative medicine approaches.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Culture Media , Endothelial Cells , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Proteomics , Reproducibility of Results
9.
J Cell Physiol ; 237(1): 675-686, 2022 01.
Article in English | MEDLINE | ID: mdl-34368976

ABSTRACT

Tendon and ligament (T/L) engineering strategies towards clinical practice have been challenged by a paucity of understanding in the identification and still poorly described characterization of cellular niches. Prospecting how resident cell populations behave in vitro, and how cryopreservation may influence T/ L-promoting factors, can provide insights into T/ L-cellular profiles for novel regenerative solutions. Therefore, we studied human T/ L-derived cells isolated from patellar tendons and cruciate ligaments as suitable cellular models to anticipate tendon and ligament niches responses for advanced strategies with predictive tenogenic and ligamentogenic value. Our results show that the crude populations isolated from tendon and ligament tissues hold a stem cell subset and share a similar behavior in terms of tenogenic/ligamentogenic commitment. Both T/ L-derived cells successfully undergo cryopreservation/thawing maintaining the tenogenic/ligamentogenic profiles. The major differences between cryopreserved and fresh populations were observed at the gene expression of MKX, SCX, and TNMD as well as at the protein levels of collagen type I and III, in which cells from tendon origin (hTDCs) evidence increased values in comparison to the ones from ligament (hLDCs, p < 0.05). In addition, low-temperature storage was shown to potentiate an immunomodulatory profile of cells, especially in hTDCs leading to an increase in the gene expression of the anti-inflammatory factors IL-4 and IL-10 (p < 0.05), as well as in the protein secretion of IL-10 (p < 0.01) and IL-4 (p < 0.001). Overall, the outcomes highlight the relevance of the cryopreserved T/ L-derived cells and their promising immunomodulatory cues as in vitro models for investigating cell-mediated mechanisms driving tissue healing and regeneration.


Subject(s)
Interleukin-10 , Interleukin-4 , Cell Differentiation , Cryopreservation , Humans , Ligaments , Tendons
10.
Acta Biomater ; 131: 236-247, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34192569

ABSTRACT

Cell sheet technology and magnetic based tissue engineering hold the potential to become instrumental in developing magnetically responsive living tissues analogues that can be potentially used both for modeling and therapeutical purposes. Cell sheet constructions more closely recreate physiological niches, through the preservation of contiguous cells and cell-ECM interactions, which assist the cellular guidance in regenerative processes. We herein propose to use magnetically assisted cell sheets (magCSs) constructed with human tendon-derived cells (hTDCs) and magnetic nanoparticles to study inflammation activity upon magCSs exposure to IL-1ß, anticipating its added value for tendon disease modeling. Our results show that IL-1ß induces an inflammatory profile in magCSs, supporting its in vitro use to enlighten inflammation mediated events in tendon cells. Moreover, the response of magCSs to IL-1ß is modulated by pulsed electromagnetic field (PEMF) stimulation, favoring the expression of anti-inflammatory genes, which seems to be associated to MAPK(ERK1/2) pathway. The anti-inflammatory response to PEMF together with the immunomodulatory potential of magCSs opens new perspectives for their applicability on tendon regeneration that goes beyond advanced cell based modeling. STATEMENT OF SIGNIFICANCE: The combination of cell sheets and magnetic-based technologies holds promise as instrumental bio-instructive tools both for tendon disease modelling and for the development of magnetically responsive living tendon substitutes. We have previously shown that remote actuation of a pulsed electromagnetic field (PEMF) modulated the inflammatory response of IL-1ß-treated human tendon-derived cell (hTDCs) monolayers. As magnetic cell sheets (magCSs) technologies enable improved cellular organization and matrix deposition, these constructions could better recapitulate tendon niches. In this work, we aimed to apply magCSs technologies to study hTDCs responses in inflammatory environments. Overall results show that PEMF-stimulated-magCSs hold evidence for immunomodulatory properties and to become a living tendon model envisioning tendon regenerative therapies.


Subject(s)
Tendons , Tissue Engineering , Cell Communication , Electromagnetic Fields , Humans , Magnetics
11.
J Mater Chem B ; 9(5): 1259-1271, 2021 02 07.
Article in English | MEDLINE | ID: mdl-33410453

ABSTRACT

In recent years, the inputs from magnetically assisted strategies have been contributing to the development of more sensitive screening methods and precise means of diagnosis to overcome existing and emerging treatment challenges. The features of magnetic materials enabling in vivo traceability, specific targeting and space- and time-controlled delivery of nanomedicines have highlighted the resourcefulness of the magnetic toolbox for biomedical applications and theranostic strategies. The breakthroughs in magnetically assisted technologies for contact-free control of cell and tissue fate opens new perspectives to improve healing and instruct regeneration reaching a wide range of diseases and disorders. In this review, the contribution of magnetic nanoparticles (MNPs) will be explored as sophisticated and versatile nanotriggers, evidencing their unique cues to probe and control cell function. As cells detect and engage external magnetic features, these approaches will be overviewed considering molecular engineering and cell programming perspectives as well as cell and tissue targeting modalities. The therapeutic relevance of MNPs will be also emphasized as key components of nanostructured systems to control the release of nanomedicines and in the context of new therapy technologies.


Subject(s)
Biosensing Techniques/methods , Magnetic Phenomena , Magnetics/methods , Humans
12.
ACS Appl Bio Mater ; 4(8): 6023-6035, 2021 08 16.
Article in English | MEDLINE | ID: mdl-35006866

ABSTRACT

Tissue engineered (TE) substitutes of clinically relevant sizes need an adequate vascular system to ensure function and proper tissue integration after implantation. However, the predictable vascularization of TE substitutes is yet to be achieved. Molecular weight variations in hyaluronic acid (HA) have been pointed to trigger angiogenesis. Thus, this study investigates HA oligomer immobilization as a promoter for TE construct vascularization. As a proof-of-concept, the surface of methacrylated gelatin (GelMA) hydrogels were functionalized with high molecular weight (HMW; 1.5 to 1.8 MDa) and low molecular weight (LMW; < 10 kDa) HA, previously modified with aldehyde groups to enable the immobilization through Schiff's base formation. The ability of A-HA to bind amine-presenting surfaces was confirmed by Surface Plasmon Resonance (SPR). Human Umbilical Vein Endothelial Cells (HUVECs) seeded over hydrogels functionalized with LMW HA showed higher proliferation and expression of angiogenic markers (KDR and CD31), than those grown in HMW HA conjugated- or plain surfaces, in line with the activation of HA ERK1/2 mediated downstream signaling. Moreover, when cocultured with human dental pulp cells (hDPCs) encapsulated into the GelMA, an increase in endothelial cell migration was observed for the LMW HA functionalized formulations. Overall LMW HA functionalization enhanced endothelial cell response showing potential as an angiogenesis inducer for TE applications.


Subject(s)
Hyaluronic Acid , Tissue Engineering , Gelatin/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hyaluronic Acid/pharmacology , Hydrogels/metabolism
13.
Int J Mol Sci ; 21(15)2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32751697

ABSTRACT

Inflammation is part of the natural healing response, but it has been simultaneously associated with tendon disorders, as persistent inflammatory events contribute to physiological changes that compromise tendon functions. The cellular interactions within a niche are extremely important for healing. While human tendon cells (hTDCs) are responsible for the maintenance of tendon matrix and turnover, macrophages regulate healing switching their functional phenotype to environmental stimuli. Thus, insights on the hTDCs and macrophages interactions can provide fundamental contributions on tendon repair mechanisms and on the inflammatory inputs in tendon disorders. We explored the crosstalk between macrophages and hTDCs using co-culture approaches in which hTDCs were previously stimulated with IL-1ß. The potential modulatory effect of the pulsed electromagnetic field (PEMF) in macrophage-hTDCs communication was also investigated using the magnetic parameters identified in a previous work. The PEMF influences a macrophage pro-regenerative phenotype and favors the synthesis of anti-inflammatory mediators. These outcomes observed in cell contact co-cultures may be mediated by FAK signaling. The impact of the PEMF overcomes the effect of IL-1ß-treated-hTDCs, supporting PEMF immunomodulatory actions on macrophages. This work highlights the relevance of intercellular communication in tendon healing and the beneficial role of the PEMF in guiding inflammatory responses toward regenerative strategies.


Subject(s)
Cell Communication/genetics , Inflammation/genetics , Interleukin-1beta/genetics , Macrophage Activation/genetics , Cell Communication/radiation effects , Cell Polarity/genetics , Cell Polarity/radiation effects , Coculture Techniques , Electromagnetic Fields , Humans , Inflammation/immunology , Inflammation/therapy , Macrophages/immunology , Macrophages/metabolism , Magnetic Field Therapy , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/radiation effects , Signal Transduction , Tendon Injuries/genetics , Tendon Injuries/pathology , Tendon Injuries/therapy , Tendons/metabolism , Tendons/pathology , Tendons/radiation effects , Tumor Necrosis Factor-alpha/genetics , Wound Healing/genetics , Wound Healing/radiation effects
14.
Acta Biomater ; 113: 488-500, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32652226

ABSTRACT

Injuries affecting load bearing tendon tissues are a significant clinical burden and efficient treatments are still unmet. Tackling tendon regeneration, tissue engineering strategies aim to develop functional substitutes that recreate native tendon milieu. Tendon mimetic scaffolds capable of remote magnetic responsiveness and functionalized magnetic nanoparticles (MNPs) targeting cellular mechanosensitive receptors are potential instructive tools to mediate mechanotransduction in guiding tenogenic responses. In this work, we combine magnetically responsive scaffolds and targeted Activin A type II receptor in human adipose stem cells (hASCs), under alternating magnetic field (AMF), to synergistically facilitate external control over signal transduction. The combination of remote triggering TGF-ß/Smad2/3 using MNPs tagged hASCs, through magnetically actuated scaffolds, stimulates overall expression of tendon related genes and the deposition of tendon related proteins, in comparison to non-stimulated conditions. Moreover, the phosphorylation of Smad2/3 proteins and their nuclear co-localization was also more evident. Overall, biophysical stimuli resulting from magnetic scaffolds and magnetically triggered cells under AMF stimulation modulate the mechanosensing response of hASCs towards tenogenesis, holding therapeutic promise. STATEMENT OF SIGNIFICANCE: The concept of magnetically-assisted tissue engineering may assist the development of innovative solutions to treat tendon disorders upon remote control of biological processes as cell migration or differentiation. Herein, we originally combine a fibrous aligned superparamagnetic scaffold, based on a biodegradable polymeric blend of starch and poly-ɛ-caprolactone incorporating magnetic nanoparticles (MNPs), and human adipose stem cells (hASCs) labelled with MNPs functionalized with anti-activin receptor type IIA (ActRIIA). Constructs were stimulated using alternating magnetic field (AMF), to activate the ActRIIA and subsequent induction of TGF-ß signaling, through Smad2/3 phosphorylation cascade, enhancing the expression of tendon-related markers. Altogether, these findings contribute with powerful bio-magnetic approaches to activate key tenogenic pathways, envisioning future translation of magnetic biomaterials into regenerative platforms for tendon repair.


Subject(s)
Biological Phenomena , Mechanotransduction, Cellular , Adipose Tissue , Cell Differentiation , Humans , Magnetic Phenomena , Signal Transduction , Smad2 Protein , Stem Cells , Tissue Engineering , Tissue Scaffolds
15.
J Orthop Res ; 38(1): 160-172, 2020 01.
Article in English | MEDLINE | ID: mdl-31769535

ABSTRACT

Strategies aiming at controlling and modulating inflammatory cues may offer therapeutic solutions for improving tendon regeneration. This study aims to investigate the modulatory effect of pulsed electromagnetic field (PEMF) on the inflammatory profile of human tendon-derived cells (hTDCs) after supplementation with interleukin-1ß (IL-1ß). IL-1ß was used to artificially induce inflammatory cues associated with injured tendon environments. The PEMF effect was investigated varying the frequency (5 or 17 Hz), intensity (1.5, 4, or 5 mT), and duty-cycle (10% or 50%) parameters to which IL-1ß-treated hTDCs were exposed to. A PEMF actuation with 4 mT, 5 Hz and a 50% duty cycle decreased the production of IL-6 and tumor necrosis factor-α (TNF-α), as well as the expression of TNFα, IL-6, IL-8, COX-2, MMP-1, MMP-2, and MMP-3, while IL-4, IL-10, and TIMP-1 expression increased. These results suggest that PEMF stimulation can modulate hTDCs response in an inflammatory environment holding therapeutic potential for tendon regenerative strategies. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:160-172, 2020.


Subject(s)
Electromagnetic Fields , Interleukin-1beta/pharmacology , Tendons/cytology , Adult , Cell Communication/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interleukin-6/metabolism , MAP Kinase Signaling System , Tendons/immunology , Tumor Necrosis Factor-alpha/metabolism
16.
J Tissue Eng Regen Med ; 13(12): 2204-2217, 2019 12.
Article in English | MEDLINE | ID: mdl-31606945

ABSTRACT

Identification of a suitable cell source and bioactive agents guiding cell differentiation towards tenogenic phenotype represents a prerequisite for advancement of cell-based therapies for tendon repair. Human adipose-derived stem cells (hASCs) are a promising, yet intrinsically heterogenous population with diversified differentiation capacities. In this work, we investigated antigenically-defined subsets of hASCs expressing markers related to tendon phenotype or associated with pluripotency that might be more prone to tenogenic differentiation, when compared to unsorted hASCs. Subpopulations positive for tenomodulin (TNMD+ hASCs) and stage specific early antigen 4 (SSEA-4+ hASCs), as well as unsorted ASCs were cultured up to 21 days in basic medium or media supplemented with TGF-ß3 (10 ng/ml), or GDF-5 (50 ng/ml). Cell response was evaluated by analysis of expression of tendon-related markers at gene level and protein level by real time RT-PCR, western blot, and immunocytochemistry. A significant upregulation of scleraxis was observed for both subpopulations and unsorted hASCs in the presence of TGF-ß3. More prominent alterations in gene expression profile in response to TGF-ß3 were observed for TNMD+ hASCs. Subpopulations evidenced an increased collagen III and TNC deposition in basal medium conditions in comparison with unsorted hASCs. In the particular case of TNMD+ hASCs, GDF-5 seems to influence more the deposition of TNC. Within hASCs populations, discrete subsets could be distinguished offering varied sensitivity to specific biochemical stimulation leading to differential expression of tenogenic components suggesting that cell subsets may have distinctive roles in the complex biological responses leading to tenogenic commitment to be further explored in cell based strategies for tendon tissues.


Subject(s)
Adipose Tissue/metabolism , Cell Differentiation , Pluripotent Stem Cells/metabolism , Tendons/metabolism , Adipose Tissue/cytology , Adult , Antigens, Differentiation , Female , Humans , Pluripotent Stem Cells/cytology , Tendons/cytology
17.
Acta Biomater ; 99: 236-246, 2019 11.
Article in English | MEDLINE | ID: mdl-31505301

ABSTRACT

Microbial infections from post-surgery or other medical-related procedure is a serious health problem. Nowadays, the research is focused on the development of new drug-free materials with antibacterial properties to prevent or minimize the risk of infections. Spider silk is known for its unique biomechanical properties allied with biocompatibility. Recombinant DNA technology allows to bioengineering spider silk with antimicrobial peptides (AMP). Thus, our goal was to bioengineered spider silk proteins with AMP (6mer-HNP1) as an antibacterial drug-free coating for commercial silk sutures (Perma-Hand®) for decreasing bacterial infections. Perma-Hand® sutures were coated with 6mer-HNP1 by dip coating. In vitro tests, using human fetal lung fibroblasts (MRC5), showed that coated sutures sustained cell viability, and also, the contact with red blood cells (RBCs) demonstrate blood compatibility. Also, the coatings inhibited significantly the adherence and formation of biofilm, where sutures coated with 6mer-HNP1 produced a 1.5 log reduction of Methicillin-Resistant Staphylococcus aureus (MRSA) and a 2 log reduction of Escherichia coli (E. coli) compared to the uncoated Perma-Hand® suture. The mechanical properties of Perma-Hand® sutures were not affected by the presence of bioengineered spider silk proteins. Thus, the present work demonstrated that using spider silk drug-free coatings it is possible to improve the antibacterial properties of the commercial sutures. Furthermore, a new class of drug-free sutures for reducing post-implantation infections can be developed. STATEMENT OF SIGNIFICANCE: Microbial infections from post-surgery or other medical-related procedure is a serious health problem. Developing new drug-free materials with antibacterial properties is an approach to prevent or minimize the risk of infections. Spider silk is known for its unique biomechanical properties allied with biocompatibility. Recombinant DNA technology allow to bioengineering spider silk with antimicrobial peptides (AMP). Our goal is bioengineered spider silk proteins with AMP as an antibacterial coating for silk sutures. The coatings showed exceptional antibacterial properties and maintained intrinsic mechanical features. In vitro studies showed a positive effect of the coated sutures on the cell behavior. With this new drug-free bioengineered spider silk coating is possible to develop a new class of drug-free sutures for reducing post-implantation infections.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Infections/prevention & control , Coated Materials, Biocompatible/chemistry , Silk/chemistry , Sutures/adverse effects , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Biofilms , Biomechanical Phenomena , Coated Materials, Biocompatible/pharmacology , Escherichia coli/drug effects , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Silk/pharmacology , Spiders , Surface Properties , Surgical Wound Infection/prevention & control , Tensile Strength , alpha-Defensins/metabolism
18.
Mater Sci Eng C Mater Biol Appl ; 93: 1090-1103, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30274040

ABSTRACT

The growing interest on polymeric delivery systems for pulmonary administration of drugs anticipates a more direct and efficient treatment of diseases such as tuberculosis (TB) that uses the pulmonary route as the natural route of infection. Polymeric microparticles or nano-in-microparticles offer target delivery of drugs to the lungs and the potential to control and sustain drug release within TB infected macrophages improving the efficiency of the anti-TB treatment and reducing side effects. In a dry powder form these inhalable delivery systems have increased stability and prolonged storage time without requiring refrigeration, besides being cost-effective and patient convenient. Thus, this review aims to compile the recent innovations of inhalable polymeric dry powder systems for the delivery of anti-TB drugs exploring the methods of production, aerodynamic characterization and the efficacy of targeted drug delivery systems using in vitro and in vivo models of the disease. Advanced knowledge and promising outcomes of these systems are anticipated to simplify and revolutionize the pulmonary drug delivery and to contribute towards more effective anti-TB treatments.


Subject(s)
Antitubercular Agents/therapeutic use , Drug Delivery Systems/methods , Tuberculosis/drug therapy , Administration, Inhalation , Animals , Humans , Powders
19.
J Mater Sci Mater Med ; 29(6): 88, 2018 Jun 14.
Article in English | MEDLINE | ID: mdl-29904797

ABSTRACT

The restoration of dentine-pulp complex remains a challenge for dentists; nonetheless, it has been poorly addressed. An ideal system should modulate the host response, as well as enable the recruitment, proliferation and differentiation of relevant progenitor cells. Herein was proposed a photocrosslinkable hydrogel system based on hyaluronic acid (HA) and platelet lysate (PL). PL is a cocktail of growth factors (GFs) and cytokines involved in wound healing orchestration, obtained by the cryogenic processing of platelet concentrates, and was expected to provide the HA hydrogels specific biochemical cues to enhance pulp cells' recruitment, proliferation and differentiation. Stable HA hydrogels incorporating PL (HAPL) were prepared after photocrosslinking of methacrylated HA (Met-HA) previously dissolved in PL, triggered by the Ultra Violet activated photoinitiator Irgacure 2959. Both the HAPL and plain HA hydrogels were shown to be able to recruit cells from a cell monolayer of human dental pulp stem cells (hDPSCs) isolated from permanent teeth. The hDPCs were also seeded directly over the hydrogels (5 × 104 cells/hydrogel) and cultured in osteogenic conditions. Cell metabolism and DNA quantification were higher, in all time-points, for PL supplemented hydrogels (p < 0,05). Alkaline phosphatase (ALPL) activity and calcium quantification peaks were observed for the HAPL group at 21 days (p < 0,05). The gene expression for ALPL and COLIA1 was up-regulated at 21 days to HAPL, compared with HA group (p < 0,05). Within the same time point, the gene expression for RUNX2 did not differ between the groups. Overall, data demonstrated that the HA hydrogels incorporating PL increased the cellular metabolism and stimulate the mineralized matrix deposition by hDPSCs, providing clear evidence of the potential of the proposed system for the repair of damaged pulp/dentin tissue and endodontics regeneration.


Subject(s)
Blood Platelets/cytology , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Stem Cells/cytology , Alkaline Phosphatase/metabolism , Calcium/chemistry , Cell Differentiation , Cell Proliferation , Chemotaxis , Cross-Linking Reagents/chemistry , Dental Pulp/cytology , Flow Cytometry , Gene Expression Profiling , Humans , Osteogenesis , Photochemistry , Regeneration , Tissue Engineering , Tooth/cytology
20.
ACS Appl Mater Interfaces ; 10(27): 23311-23320, 2018 Jul 11.
Article in English | MEDLINE | ID: mdl-29906095

ABSTRACT

The present work has explored bioactive glass nanoparticles (BGNPs) and developed strontium-doped nanoparticles (BGNPsSr), envisioning orthopedic strategies compatible with vascularization. The nanoparticles were synthesized by the sol-gel method, achieving a diameter of 55 nm for BGNPs and 75 nm for BGNPsSr, and the inclusion of strontium caused no structural alteration. The nanoparticles exhibited high cytocompatibility for human umbilical vein endothelial cells (HUVECs) and SaOS-2. Additionally, the incorporation of strontium emphasized the tubule networking behavior of HUVECs. Our results demonstrate that the nanoparticle dissolution products encouraged the osteogenic differentiation of human adipose stem cells as it favored the expression of key genes and proteins associated with osteogenic lineage. This effect was markedly enhanced for BGNPsSr, which could prompt stem cell osteogenic differentiation without the typical osteogenic inducers. This study not only supports the hypothesis that BGNPs might play a significant role in osteogenic commitment but also highlights that the designed BGNPsSr is a valuable tool for stem cell "tune-up" in bone tissue engineering applications.

SELECTION OF CITATIONS
SEARCH DETAIL
...