Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Adv Exp Med Biol ; 2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38904750

RESUMEN

Stem cells have been attractive targets for tissue regeneration due to their inherent ability to differentiate into various specialized cell types; however, efforts for stem cell transplantation for the treatment of degenerated tissues have been hampered by the propensity of some stem cell types to form teratomas and the lessened viability of stem cells after transplantation. These disadvantages can be prevented using tailored extracellular matrix-like materials that can be used as an aid for the transplantation of stem cells. Nanomaterials, in particular nanofibers, have great potential to be used as extracellular matrix-like materials for this purpose. In this article, we will review the use of stem cells and nanofibers for skin regeneration and wound healing.

2.
Biomater Sci ; 11(14): 5012-5024, 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37334774

RESUMEN

Peptide amphiphiles (PAs) have emerged as effective molecular building blocks for creating self-assembling nanobiomaterials for multiple biomedical applications. Herein, we report a straightforward approach to assemble soft bioinstructive platforms to recreate the native neural extracellular matrix (ECM) aiming for neuronal regeneration based on the electrostatic-driven supramolecular presentation of laminin-derived IKVAV-containing self-assembling PA (IKVAV-PA) on biocompatible multilayered nanoassemblies. Spectroscopic and microscopic techniques show that the co-assembly of positively charged low-molecular-weight IKVAV-PA with oppositely charged high-molecular-weight hyaluronic acid (HA) triggers the formation of ordered ß-sheet structures denoting a one-dimensional nanofibrous network. The successful functionalization of poly(L-lysine)/HA layer-by-layer nanofilms with an outer positively charged layer of self-assembling IKVAV-PA is demonstrated by the quartz crystal microbalance with dissipation monitoring and the nanofibrous morphological properties revealed by atomic force microscopy. The bioactive ECM-mimetic supramolecular nanofilms promote the enhancement of primary neuronal cells' adhesion, viability, and morphology when compared to the PA without the IKVAV sequence and PA-free biopolymeric multilayered nanofilms, and stimulate neurite outgrowth. The nanofilms hold great promise as bioinstructive platforms for enabling the assembly of customized and robust multicomponent supramolecular biomaterials for neural tissue regeneration.


Asunto(s)
Matriz Extracelular , Péptidos , Péptidos/farmacología , Péptidos/química , Matriz Extracelular/química , Neuronas , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/análisis , Proyección Neuronal
3.
Pharmaceutics ; 15(4)2023 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-37111749

RESUMEN

Ranibizumab is a recombinant VEGF-A antibody used to treat the wet form of age-related macular degeneration. It is intravitreally administered to ocular compartments, and the treatment requires frequent injections, which may cause complications and patient discomfort. To reduce the number of injections, alternative treatment strategies based on relatively non-invasive ranibizumab delivery are desired for more effective and sustained release in the eye vitreous than the current clinical practice. Here, we present self-assembled hydrogels composed of peptide amphiphile molecules for the sustained release of ranibizumab, enabling local high-dose treatment. Peptide amphiphile molecules self-assemble into biodegradable supramolecular filaments in the presence of electrolytes without the need for a curing agent and enable ease of use due to their injectable nature-a feature provided by shear thinning properties. In this study, the release profile of ranibizumab was evaluated by using different peptide-based hydrogels at varying concentrations for improved treatment of the wet form of age-related macular degeneration. We observed that the slow release of ranibizumab from the hydrogel system follows extended- and sustainable release patterns without any dose dumping. Moreover, the released drug was biologically functional and effective in blocking the angiogenesis of human endothelial cells in a dose-dependent manner. In addition, an in vivo study shows that the drug released from the hydrogel nanofiber system can stay in the rabbit eye's posterior chamber for longer than a control group that received only a drug injection. The tunable physiochemical characteristics, injectable nature, and biodegradable and biocompatible features of the peptide-based hydrogel nanofiber show that this delivery system has promising potential for intravitreal anti-VEGF drug delivery in clinics to treat the wet form age-related macular degeneration.

4.
Regen Biomater ; 10: rbac084, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36683737

RESUMEN

Articular cartilage, which is exposed to continuous repetitive compressive stress, has limited self-healing capacity in the case of trauma. Thus, it is crucial to develop new treatment options for the effective regeneration of the cartilage tissue. Current cellular therapy treatment options are microfracture and autologous chondrocyte implantation; however, these treatments induce the formation of fibrous cartilage, which degenerates over time, rather than functional hyaline cartilage tissue. Tissue engineering studies using biodegradable scaffolds and autologous cells are vital for developing an effective long-term treatment option. 3D scaffolds composed of glycosaminoglycan-like peptide nanofibers are synthetic, bioactive, biocompatible, and biodegradable and trigger cell-cell interactions that enhance chondrogenic differentiation of cells without using any growth factors. We showed differentiation of mesenchymal stem cells into chondrocytes in both 2D and 3D culture, which produce a functional cartilage extracellular matrix, employing bioactive cues integrated into the peptide nanofiber scaffold without adding exogenous growth factors.

5.
Macromol Biosci ; 21(1): e2000234, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33043585

RESUMEN

The highly complex nature of spinal cord injuries (SCIs) requires design of novel biomaterials that can stimulate cellular regeneration and functional recovery. Promising SCI treatments use biomaterial scaffolds, which provide bioactive cues to the cells in order to trigger neural regeneration in the spinal cord. In this work, the use of peptide nanofibers is demonstrated, presenting protein binding and cellular adhesion epitopes in a rat model of SCI. The self-assembling peptide molecules are designed to form nanofibers, which display heparan sulfate mimetic and laminin mimetic epitopes to the cells in the spinal cord. These neuroactive nanofibers are found to support adhesion and viability of dorsal root ganglion neurons as well as neurite outgrowth in vitro and enhance tissue integrity after 6 weeks of injury in vivo. Treatment with the peptide nanofiber scaffolds also show significant behavioral improvement. These results demonstrate that it is possible to facilitate regeneration especially in the white matter of the spinal cord, which is usually damaged during the accidents using bioactive 3D nanostructures displaying high densities of laminin and heparan sulfate-mimetic epitopes on their surfaces.


Asunto(s)
Nanofibras/química , Péptidos/farmacología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Médula Espinal/efectos de los fármacos , Sustancia Blanca/crecimiento & desarrollo , Animales , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Epítopos/efectos de los fármacos , Ganglios Espinales/efectos de los fármacos , Humanos , Regeneración Nerviosa/efectos de los fármacos , Neuronas/efectos de los fármacos , Péptidos/química , Unión Proteica/efectos de los fármacos , Ratas , Médula Espinal/crecimiento & desarrollo , Médula Espinal/patología , Traumatismos de la Médula Espinal/patología , Sustancia Blanca/efectos de los fármacos
6.
Biotechnol J ; 15(12): e2000100, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32679620

RESUMEN

Biotinylated peptide amphiphile (Biotin-PA) nanofibers, are designed as a noncovalent binding location for antigens, which are adjuvants to enhance, accelerate, and prolong the immune response triggered by antigens. Presenting antigens on synthetic Biotin-PA nanofibers generated a higher immune response than the free antigens delivered with a cytosine-phosphate-guanine oligodeoxynucleotides (CpG ODN) (TLR9 agonist) adjuvant. Antigen attached Biotin-PA nanofibers trigger splenocytes to produce high levels of cytokines (IFN-γ, IL-12, TNF-α, and IL-6) and to exhibit a superior cross-presentation of the antigen. Both Biotin-PA nanofibers and CpG ODN induce a Th-1-biased IgG subclass response; however, delivering the antigen with Biotin-PA nanofibers induce significantly greater production of total IgG and subclasses of IgG compared to delivering the antigen with CpG ODN. Contrary to CpG ODN, Biotin-PA nanofibers also enhance antigen-specific splenocyte proliferation and increase the proportion of the antigen-specific CD8(+) T cells. Given their biodegradability and biocompatibility, Biotin-PA nanofibers have a significant potential in immunoengineering applications as a biomaterial for the delivery of a diverse set of antigens derived from intracellular pathogens, emerging viral diseases such as COVID-19, or cancer cells to induce humoral and cellular immune responses against the antigens.


Asunto(s)
Adyuvantes Inmunológicos/química , Nanofibras/química , Péptidos/química , Péptidos/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Presentación de Antígeno , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/inmunología , Antígenos/administración & dosificación , Antígenos/química , Materiales Biocompatibles/química , Biotecnología , Biotina/análogos & derivados , Citocinas/metabolismo , Diseño de Fármacos , Inmunidad Celular , Inmunidad Humoral , Técnicas In Vitro , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nanofibras/administración & dosificación , Nanofibras/ultraestructura , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Péptidos/administración & dosificación , Ingeniería de Proteínas
7.
Bioconjug Chem ; 30(9): 2417-2426, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31415164

RESUMEN

Cadherins are vital for cell-to-cell interactions during tissue growth, migration, and differentiation processes. Both biophysical and biochemical inputs are generated upon cell-to-cell adhesions, which determine the fate of the mesenchymal stem cells (MSCs). The effect of cadherin interactions on the MSC differentiation still remains elusive. Here we combined the N-Cadherin mimetic peptide (HAV-PA) with the self-assembling E-PA and the resultant N-cadherin mimetic peptide nanofibers promoted chondrogenic differentiation of MSCs in conjunction with chondrogenic factors as a synthetic extracellular matrix system. Self-assembly of the precursor peptide amphiphile molecules HAV-PA and E-PA enable the organization of HAV peptide residues in close proximity to the cell interaction site, forming a supramolecular N-cadherin-like system. These bioactive peptide nanofibers not only promoted viability and enhanced adhesion of MSCs but also augmented the expression of cartilage specific matrix components compared to the nonbioactive control nanofibers. Overall, the N-cadherin mimetic peptide nanofiber system facilitated MSC commitment into the chondrogenic lineage presenting an alternative bioactive platform for stem-cell-based cartilage regeneration.


Asunto(s)
Cadherinas/química , Diferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Nanofibras/química , Peptidomiméticos/química , Peptidomiméticos/farmacología , Secuencia de Aminoácidos , Animales , Interacciones Hidrofóbicas e Hidrofílicas , Células Madre Mesenquimatosas/efectos de los fármacos , Ratas
8.
Eur Phys J E Soft Matter ; 42(5): 63, 2019 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-31115713

RESUMEN

Supramolecular peptide nanofibers that are composed of peptide amphiphile molecules have been widely used for many purposes from biomedical applications to energy conversion. The self-assembly mechanisms of these peptide nanofibers also provide convenient models for understanding the self-assembly mechanisms of various biological supramolecular systems; however, the current theoretical models that explain these mechanisms do not sufficiently explain the experimental results. In this study, we present a new way of modeling these nanofibers that better fits with the experimental data. Molecular dynamics simulations were applied to create model fibers using two different layer models and two different tilt angles. Strikingly, the fibers which were modeled to be tilting the peptide amphiphile molecules and/or tilting the plane were found to be more stable and consistent with the experiments.

9.
ACS Appl Bio Mater ; 2(4): 1686-1695, 2019 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35026903

RESUMEN

Lower back pain (LBP) is a prevalent spinal symptom at the lumbar region of the spine, which severely effects quality of life and constitutes the number one cause of occupational disability. Degeneration of the intervertebral disc (IVD) is one of the well-known causes contributing to the LBP. Therapeutic biomaterials inducing IVD regeneration are promising candidates for IVD degeneration treatments. Here, we demonstrate a collagen peptide presenting nanofiber scaffold to mimic the structure and function of the natural extracellular matrix of the tissue for IVD regeneration. The collagen peptide presenting nanofiber was designed by using a Pro-Hyp-Gly (POG) peptide sequence on a self-assembling peptide amphiphile molecule, which assembled into nanofibers forming scaffolds. Injection of collagen peptide presenting peptide nanofiber scaffold into the degenerated rabbit IVDs induced more glycosaminoglycan and collagen deposition compared to controls. Functional recovery of the tissue was evaluated by degeneration index score, where the bioactive scaffold was shown to provide functional recovery of the IVD degeneration. These results showed that the collagen peptide presenting nanofiber scaffold can prevent the progression of IVD degeneration and provide further functional recovery of the tissue.

10.
Macromol Biosci ; 19(1): e1800080, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29745025

RESUMEN

Dentin phosphoprotein (DPP) is a major component of the dentin matrix playing crucial role in hydroxyapatite deposition during bone mineralization, making it a prime candidate for the design of novel materials for bone and tooth regeneration. The bioactivity of DPP-derived proteins is controlled by the phosphorylation and dephosphorylation of the serine residues. Here an enzyme-responsive peptide nanofiber system inducing biomineralization is demonstrated. It closely emulates the structural and functional properties of DPP and facilitates apatite-like mineral deposition. The DPP-mimetic peptide molecules self-assemble through dephosphorylation by alkaline phosphatase (ALP), an enzyme participating in tooth and bone matrix mineralization. Nanofiber network formation is also induced through addition of calcium ions. The gelation process following nanofiber formation produces a mineralized extracellular matrix like material, where scaffold properties and phosphate groups promote mineralization. It is demonstrated that the DPP-mimetic peptide nanofiber networks can be used for apatite-like mineral deposition for bone regeneration.


Asunto(s)
Materiales Biomiméticos , Regeneración Ósea/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Proteínas de la Matriz Extracelular , Nanofibras/química , Péptidos , Fosfoproteínas , Sialoglicoproteínas , Materiales Biomiméticos/síntesis química , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/farmacología , Humanos , Péptidos/síntesis química , Péptidos/química , Péptidos/farmacología , Fosfoproteínas/química , Fosfoproteínas/farmacología , Sialoglicoproteínas/química , Sialoglicoproteínas/farmacología
11.
Biomater Sci ; 6(7): 1859-1868, 2018 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-29799029

RESUMEN

The development of new biomaterials mimicking the neuronal extracellular matrix (ECM) requires signals for the induction of neuronal differentiation and regeneration. In addition to the biological and chemical cues, the physical properties of the ECM should also be considered while designing regenerative materials for nervous tissue. In this study, we investigated the influence of the microenvironment on tenascin-C signaling using 2D surfaces and 3D scaffolds generated by a peptide amphiphile nanofiber gel with a tenascin-C derived peptide epitope (VFDNFVLK). While tenascin-C mimetic PA nanofibers significantly increased the length and number of neurites produced by PC12 cells on 2D cell culture, more extensive neurite outgrowth was observed in the 3D gel environment. PC12 cells encapsulated within the 3D tenascin-C mimetic peptide nanofiber gel also exhibited significantly increased expression of neural markers compared to the cells on 2D surfaces. Our results emphasize the synergistic effects of the 3D conformation of peptide nanofibers along with the tenascin-C signaling and growth factors on the neuronal differentiation of PC12 cells, which may further provide more tissue-like morphology for therapeutic applications.


Asunto(s)
Materiales Biomiméticos/farmacología , Nanofibras/química , Neuronas/efectos de los fármacos , Péptidos/farmacología , Transducción de Señal , Andamios del Tejido , Animales , Biomarcadores/metabolismo , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Geles , Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/farmacología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proyección Neuronal/efectos de los fármacos , Proyección Neuronal/fisiología , Neuronas/citología , Neuronas/metabolismo , Células PC12 , Péptidos/síntesis química , Ratas , Tenascina/metabolismo , Tenascina/farmacología
12.
Biomater Sci ; 6(7): 1777-1790, 2018 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-29770392

RESUMEN

Promotion of neurite outgrowth is an important limiting step for regeneration in nerve injury and depends strongly on the local expression of nerve growth factor (NGF). The rational design of bioactive materials is a promising approach for the development of novel therapeutic methods for nerve regeneration, and biomaterials capable of presenting NGF to nerve cells are especially suitable for this purpose. In this study, we show bioactive peptide amphiphile (PA) nanofibers capable of promoting neurite outgrowth by displaying high density binding epitopes for NGF. A high-affinity NGF-binding sequence was identified by phage display and combined with a beta-sheet forming motif to produce a self-assembling PA molecule. The bioactive nanofiber had higher affinity for NGF compared to control nanofibers and in vitro studies revealed that the NGF binding peptide amphiphile nanofibers (NGFB-PA nanofiber) significantly promote the neurite outgrowth of PC-12 cells. In addition, the nanofibers induced differentiation of PC-12 cells into neuron-like cells by enhancing NGF/high-activity NGF receptor (TrkA) interactions and activating MAPK pathway elements. The NGFB-PA nanofiber was further shown as a promising material to support axonal outgrowth from primary sensory neurons. These materials will pave the way for the development of new therapeutic agents for peripheral nervous system injuries.


Asunto(s)
Ganglios Espinales/efectos de los fármacos , Nanofibras/química , Proyección Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Diferenciación Celular/efectos de los fármacos , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Nanofibras/ultraestructura , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Células PC12 , Biblioteca de Péptidos , Péptidos/síntesis química , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptor trkA/genética , Receptor trkA/metabolismo
13.
Nanotechnology ; 29(28): 285701, 2018 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-29664418

RESUMEN

Biological feedback mechanisms exert precise control over the initiation and termination of molecular self-assembly in response to environmental stimuli, while minimizing the formation and propagation of defects through self-repair processes. Peptide amphiphile (PA) molecules can self-assemble at physiological conditions to form supramolecular nanostructures that structurally and functionally resemble the nanofibrous proteins of the extracellular matrix, and their ability to reconfigure themselves in response to external stimuli is crucial for the design of intelligent biomaterials systems. Here, we investigated real-time self-assembly, deformation, and recovery of PA nanofibers in aqueous solution by using a force-stabilizing double-pass scanning atomic force microscopy imaging method to disrupt the self-assembled peptide nanofibers in a force-dependent manner. We demonstrate that nanofiber damage occurs at tip-sample interaction forces exceeding 1 nN, and the damaged fibers subsequently recover when the tip pressure is reduced. Nanofiber ends occasionally fail to reconnect following breakage and continue to grow as two individual nanofibers. Energy minimization calculations of nanofibers with increasing cross-sectional ellipticity (corresponding to varying levels of tip-induced fiber deformation) support our observations, with high-ellipticity nanofibers exhibiting lower stability compared to their non-deformed counterparts. Consequently, tip-mediated mechanical forces can provide an effective means of altering nanofiber integrity and visualizing the self-recovery of PA assemblies.

14.
Acta Biomater ; 73: 263-274, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29656073

RESUMEN

Osteoarthritis (OA) is a condition where tissue function is lost through a combination of secondary inflammation and deterioration in articular cartilage. One of the most common causes of OA is age-related tissue impairment because of wear and tear due to mechanical erosion. Hyaluronic acid-based viscoelastic supplements have been widely used for the treatment of knee injuries. However, the current formulations of hyaluronic acid are unable to provide efficient healing and recovery. Here, a nanofiber-hyaluronic acid membrane system that was prepared by using a quarter of the concentration of commercially available hyaluronic acid supplement, Hyalgan®, was used for the treatment of an osteoarthritis model, and Synvisc®, which is another commercially available hyaluronic acid containing viscoelastic supplement, was used as a control. The results show that this system provides efficient protection of arthritic cartilage tissue through the preservation of cartilage morphology with reduced osteophyte formation, protection of the subchondral region from deterioration, and maintenance of cartilage specific matrix proteins in vivo. In addition, the hybrid nanofiber membrane enabled chondrocyte encapsulation and provided a suitable culturing environment for stem cell growth in vitro. Overall, our results suggest that this hybrid nanofibrous scaffold provides a potential platform the treatment of OA. STATEMENT OF SIGNIFICANCE: Osteoarthritis is a debilitating joint disease affecting millions of people worldwide. It occurs especially in knees due to aging, sport injuries or obesity. Although hyaluronic acid-based viscoelastic supplements are widely used, there is still no effective treatment method for osteoarthritis, which necessitates surgical operation as an only choice for severe cases. Therefore, there is an urgent need for efficient therapeutics. In this study, a nanofiber-HA membrane system was developed for the efficient protection of arthritic cartilage tissue from degeneration. This hybrid nanofiber system provided superior therapeutic activity at a relatively lower concentration of hyaluronic acid than Hyalgan® and Synvisc® gels, which are currently used in clinics. This work demonstrates for the first time that this hybrid nanofiber membrane scaffold can be utilized as a potential candidate for osteoarthritis treatment.


Asunto(s)
Cartílago Articular/efectos de los fármacos , Ácido Hialurónico/administración & dosificación , Nanofibras/administración & dosificación , Osteoartritis/terapia , Péptidos/administración & dosificación , Animales , Cartílago Articular/química , Supervivencia Celular , Condrocitos/citología , Cromatografía Liquida , Dicroismo Circular , Miembro Posterior/patología , Inflamación , Masculino , Espectrometría de Masas , Células Madre Mesenquimatosas/metabolismo , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Oscilometría , Osteoartritis/fisiopatología , Ratas , Ratas Sprague-Dawley , Reología , Estrés Mecánico , Andamios del Tejido
15.
Semin Cell Dev Biol ; 73: 153-164, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28811262

RESUMEN

Atomic force microscopy is an emerging tool for investigating the biomolecular aspects of cellular interactions; however, cell and tissue analyses must frequently be performed in aqueous environment, over rough surfaces, and on complex adhesive samples that complicate the imaging process and readily facilitate the blunting or fouling of the AFM probe. In addition, the shape and surface chemistry of the probe determine the quality and types of data that can be acquired from biological materials, with certain information becoming available only within a specific range of tip lengths or diameters, or through the assistance of specific chemical or biological functionalization procedures. Consequently, a broad range of probe modification techniques has been developed to extend the capabilities and overcome the limitations of biological AFM measurements, including the fabrication of AFM tips with specialized morphologies, surface coating with biologically affine molecules, and the attachment of proteins, nucleic acids and cells to AFM probes. In this review, we underline the importance of probe choice and modification for the AFM analysis of biomaterials, discuss the recent literature on the use of non-standard AFM tips in life sciences research, and consider the future utility of tip functionalization methods for the investigation of fundamental cell and tissue interactions.


Asunto(s)
Materiales Biocompatibles/análisis , Materiales Biocompatibles/química , Microscopía de Fuerza Atómica , Animales , Humanos
16.
ACS Appl Mater Interfaces ; 10(1): 308-317, 2018 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-29232108

RESUMEN

Peripheral nerve injuries cause devastating problems for the quality of patients' lives, and regeneration following damage to the peripheral nervous system is limited depending on the degree of the damage. Use of nanobiomaterials can provide therapeutic approaches for the treatment of peripheral nerve injuries. Electroactive biomaterials, in particular, can provide a promising cure for the regeneration of nerve defects. Here, a supramolecular electroactive nanosystem with tetra(aniline) (TA)-containing peptide nanofibers was developed and utilized for nerve regeneration. Self-assembled TA-conjugated peptide nanofibers demonstrated electroactive behavior. The electroactive self-assembled peptide nanofibers formed a well-defined three-dimensional nanofiber network mimicking the extracellular matrix of the neuronal cells. Neurite outgrowth was improved on the electroactive TA nanofiber gels. The neural differentiation of PC-12 cells was more advanced on electroactive peptide nanofiber gels, and these biomaterials are promising for further use in therapeutic neural regeneration applications.


Asunto(s)
Nanofibras , Compuestos de Anilina , Animales , Materiales Biocompatibles , Diferenciación Celular , Regeneración Nerviosa , Células PC12 , Péptidos , Ratas
17.
Nanomedicine ; 14(7): 2433-2454, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-28552644

RESUMEN

One-dimensional (1D) carbon nanotubes (CNTs) and the two-dimensional (2D) graphene represent the most widely studied allotropes of carbon. Due to their unique structural, electrical, mechanical and optical properties, 1D and 2D carbon nanostructures are considered to be leading candidates for numerous applications in biomedical fields, including tissue engineering, drug delivery, bioimaging and biosensors. The biocompatibility and toxicity issues associated with these nanostructures have been a critical impediment for their use in biomedical applications. In this review, we present an overview of the various materials types, properties, functionalization strategies and characterization methods of 1D and 2D carbon nanomaterials and their derivatives in terms of their biomedical applications. In addition, we discuss various factors and mechanisms affecting their toxicity and biocompatibility.


Asunto(s)
Técnicas Biosensibles , Sistemas de Liberación de Medicamentos , Nanoestructuras/química , Nanotubos de Carbono/química , Ingeniería de Tejidos , Animales , Humanos
18.
Adv Exp Med Biol ; 1030: 155-166, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29081053

RESUMEN

Cartilaginous tissue requires structural and metabolic support after traumatic or chronic injuries because of its limited capacity for regeneration. However, current techniques for cartilage regeneration are either invasive or ineffective for long-term repair. Developing alternative approaches to regenerate cartilage tissue is needed. Therefore, versatile scaffolds formed by biomaterials are promising tools for cartilage regeneration. Bioactive scaffolds further enhance the utility in a broad range of applications including the treatment of major cartilage defects. This chapter provides an overview of cartilage tissue, tissue defects, and the methods used for regeneration, with emphasis on peptide scaffold materials that can be used to supplement or replace current medical treatment options.


Asunto(s)
Materiales Biocompatibles/química , Cartílago/fisiopatología , Péptidos/química , Regeneración , Animales , Cartílago/lesiones , Humanos , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
19.
Mol Pharm ; 14(11): 3660-3668, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-29020766

RESUMEN

Noncovalent and electrostatic interactions facilitate the formation of complex networks through molecular self-assembly in biomolecules such as proteins and glycosaminoglycans. Self-assembling peptide amphiphiles (PA) are a group of molecules that can form nanofibrous structures and may contain bioactive epitopes to interact specifically with target molecules. Here, we report the presentation of cationic peptide sequences on supramolecular nanofibers formed by self-assembling peptide amphiphiles for cooperative enhanced antibacterial activity. Antibacterial properties of self-assembled peptide nanofibers were significantly higher than soluble peptide molecules with identical amino acid sequences, suggesting that the tandem presentation of bioactive epitopes is important for designing new materials for bactericidal activity. In addition, bacteria were observed to accumulate more rapidly on peptide nanofibers compared to soluble peptides, which may further enhance antibacterial activity by increasing the number of peptide molecules interacting with the bacterial membrane. The cationic peptide amphiphile nanofibers were observed to attach to bacterial membranes and disrupt their integrity. These results demonstrate that short cationic peptides show a significant improvement in antibacterial activity when presented in the nanofiber form.


Asunto(s)
Antiinfecciosos/química , Nanofibras/química , Péptidos/química , Epítopos/química
20.
Biotechnol J ; 12(12)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28786563

RESUMEN

The extracellular matrix (ECM) provides biochemical signals and structural support for cells, and its functional imitation is a fundamental aspect of biomaterial design for regenerative medicine applications. The stimulation of neural differentiation by a laminin protein-derived epitope in two-dimensional (2D) and three-dimensional (3D) environments is investigated. The 3D gel system is found to be superior to its 2D counterpart for the induction of neural differentiation, even in the absence of a crucial biological inducer in nerve growth factor (NGF). In addition, cells cultured in 3D gels exhibits a spherical morphology that is consistent with their form under in vivo conditions. Overall, the present study underlines the impact of bioactivity, dimension, and NGF addition, as well as the cooperative effects thereof, on the neural differentiation of PC-12 cells. These results underline the significance of 3D culture systems in the development of scaffolds that closely replicate in vivo environments for the formation of cellular organoid models in vitro.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Geles/química , Laminina/química , Nanofibras/química , Neurogénesis/fisiología , Animales , Técnicas de Cultivo de Célula/instrumentación , Perfilación de la Expresión Génica , Factor de Crecimiento Nervioso/metabolismo , Neuritas/metabolismo , Células PC12 , Péptidos/química , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...