Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 643: 39-47, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36586157

RESUMEN

Innate immune cells play a pivotal role in controlling tissue repair and rejection after biomaterial implantation. Calcium supplementation regulates cellular responses and alter the pathophysiology of various diseases. A series of macrophage activations through differential plasticity has been observed after cell-to-material interactions. We investigated the role of calcium supplementation in controlling macrophage phenotypes in pro-inflammatory and pre-reparative states. Oxidative defence and mitochondria involvement in cellular plasticity and the sequential M0 to M1 and M1 to M2 transitions were observed after calcium supplementation. This study describes the molecular mechanism of reactive oxygen species and drives the interconnected cellular plasticity of macrophages in the presence of calcium. Gene expression, and immunostaining, revealed a relationship between MHC class II maturation and cellular plasticity. This study elucidated the role of controlled calcium supplementation under various conditions. These findings underscore the molecular mechanism of calcium-mediated immune induction and its favourable use in different calcium-containing biomaterials., essential for tissue regeneration.


Asunto(s)
Calcio , Monocitos , Humanos , Monocitos/metabolismo , Calcio/metabolismo , Macrófagos/metabolismo , Fenotipo , Materiales Biocompatibles/farmacología
2.
J Cell Sci ; 133(23)2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33310869

RESUMEN

Bone morphogenetic protein (BMP) signalling plays a significant role during embryonic cartilage development and has been associated with osteoarthritis (OA) pathogenesis, being in both cases involved in triggering hypertrophy. Inspired by recent findings that BMP inhibition counteracts hypertrophic differentiation of human mesenchymal progenitors, we hypothesized that selective inhibition of BMP signalling would mitigate hypertrophic features in OA cartilage. First, a 3D in vitro OA micro-cartilage model was established using minimally expanded OA chondrocytes that was reproducibly able to capture OA-like hypertrophic features. BMP signalling was then restricted by means of two BMP receptor type I inhibitors, resulting in reduction of OA hypertrophic traits while maintaining synthesis of cartilage extracellular matrix. Our findings open potential pharmacological strategies for counteracting cartilage hypertrophy in OA and support the broader perspective that key signalling pathways known from developmental processes can guide the understanding, and possibly the mitigation, of adult pathological features.


Asunto(s)
Cartílago Articular , Osteoartritis , Adulto , Proteína Morfogenética Ósea 2 , Condrocitos , Humanos , Hipertrofia , Osteoartritis/tratamiento farmacológico , Osteoartritis/genética
3.
Int J Cancer ; 145(5): 1254-1269, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31008529

RESUMEN

Recent advancement in understanding cancer etiology has highlighted epigenetic deregulation as an important phenomenon leading to poor prognosis in glioblastoma (GBM). Polycomb repressive complex 2 (PRC2) is one such important epigenetic modifier reportedly altered in GBM. However, its defined mechanism in tumorigenesis still remains elusive. In present study, we analyzed our in-house ChIPseq data for H3k27me3 modified miRNAs and identified miR-490-3p to be the most common target in GBM with significantly downregulated expression in glioma patients in both TCGA and GBM patient cohort. Our functional analysis delineates for the first time, a central role of PRC2 catalytic unit EZH2 in directly regulating expression of this miRNA and its host gene CHRM2 in GBM. In accordance, cell line treatment with EZH2 siRNA and 5-azacytidine also confirmed its coregulation by CpG and histone methylation based epigenetic mechanisms. Furthermore, induced overexpression of miR-490-3p in GBM cell lines significantly inhibited key hallmarks including cellular proliferation, colony formation and spheroid formation, as well as epithelial-to-mesenchymal transition (EMT), with downregulation of multiple EMT transcription factors and promigratory genes (MMP9, CCL5, PIK3R1, ICAM1, ADAM17 and NOTCH1). We also for the first time report TGFBR1 and TGIF2 as two direct downstream effector targets of miR-490-3p that are also deregulated in GBM. TGIF2, a novel target, was shown to promote migration and EMT that could partially be rescued by miR-490-3p overexpression. Overall, this stands as a first study that provides a direct link between epigenetic modulator EZH2 and oncogenic TGF-ß signaling involving novel miR-490-3p/TGIF2/TGFBR1 axis, that being targetable might be promising in developing new therapeutic intervention strategies for GBM.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Glioblastoma/genética , Glioblastoma/patología , Proteínas de Homeodominio/metabolismo , MicroARNs/metabolismo , Complejo Represivo Polycomb 2/genética , Proteínas Represoras/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Neoplasias Encefálicas , Línea Celular Tumoral , Movimiento Celular/fisiología , Regulación hacia Abajo , Proteína Potenciadora del Homólogo Zeste 2/genética , Epigénesis Genética , Transición Epitelial-Mesenquimal , Glioblastoma/metabolismo , Proteínas de Homeodominio/genética , Humanos , MicroARNs/genética , Complejo Represivo Polycomb 2/metabolismo , Proteínas Represoras/genética , Transducción de Señal
4.
J Cell Physiol ; 233(5): 3817-3830, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28657193

RESUMEN

Corneal scarring is the major source of permanent blindness worldwide. The complex pathophysiology of corneal scarring is not comprehensibly understood as it involves the interaction of a constellation of pro-fibrotic cytokines influencing several signaling pathways involved in corneal scar development. In the present study, an attempt has been made to generate a relatively simple in vitro corneal scar model using primary corneal keratocytes by exogenously providing an optimized dose of combination of cytokines (TGF-ß1, IL-6, and IL-8) involved in scar formation in situ. Data obtained from gene and protein expression analysis depicted enhanced ECM production with discrete expression of myofibroblast specific markers. The protein-protein interactions associated these proteins to various pathways involved in wound healing, cellular migration, and cytoskeletal remodeling justifying high relevance to in vivo scar formation. Hence the developed model can be used to acquire understanding about corneal scar pathophysiology and thus might be useful for designing the treatment modalities and efficacies for controlling scar formation.


Asunto(s)
Cicatriz/patología , Córnea/patología , Lesiones de la Cornea/patología , Citoesqueleto/patología , Cicatrización de Heridas/fisiología , Animales , Movimiento Celular/fisiología , Células Cultivadas , Cabras , Miofibroblastos/patología
5.
J Cell Physiol ; 233(11): 9015-9030, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29923313

RESUMEN

Human hair dermal papilla (DP) cells are specialized mesenchymal cells that play a pivotal role in hair regeneration and hair cycle activation. The current study aimed to first develop three-dimensional (3D) DP spheroids (DPS) with or without a silk-gelatin (SG) microenvironment, which showed enhanced DP-specific gene expression, resulting in enhanced extracellular matrix (ECM) production compared with a monolayer culture. We tested the feasibility of using this DPS model for drug screening by using minoxidil, which is a standard drug for androgenic alopecia. Minoxidil-treated DPS showed enhanced expression of growth factors and ECM proteins. Further, an attempt has been made to establish an in vitro 3D organoid model consisting of DPS encapsulated by SG hydrogel and hair follicle (HF) keratinocytes and stem cells. This HF organoid model showed the importance of structural features, cell-cell interaction, and hypoxia akin to in vivo HF. The study helped to elucidate the molecular mechanisms to stimulate cell proliferation, cell viability, and elevated expression of HF markers as well as epithelial-mesenchymal crosstalks, demonstrating high relevance to human HF biology. This simple in vitro DP organoid model system has the potential to provide significant insights into the underlying mechanisms of HF morphogenesis, distinct molecular signals relevant to different stages of the hair cycle, and hence can be used for controlled evaluation of the efficacy of new drug molecules.


Asunto(s)
Folículo Piloso/crecimiento & desarrollo , Cabello/crecimiento & desarrollo , Células Madre Mesenquimatosas/citología , Organoides/crecimiento & desarrollo , Diferenciación Celular/genética , Proliferación Celular/genética , Células Cultivadas , Dermis/citología , Dermis/crecimiento & desarrollo , Transición Epitelial-Mesenquimal/genética , Femenino , Cabello/citología , Folículo Piloso/citología , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Organoides/citología , Regeneración/genética
6.
Gut ; 70(5): 998-999, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32694173

Asunto(s)
Fallo Hepático , Humanos
7.
Lab Invest ; 96(12): 1311-1326, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27748734

RESUMEN

Macular corneal dystrophy (MCD) is characterized by multiple punctate gray-white opacities in the corneal stromal region, due to the accumulation of abnormally sulfated keratan sulfates. We attempted to develop an in vitro model of MCD by simulating the sulfation inhibition using sodium chlorate, a chemical inhibitor of 3'-phosphoadenosine-5'-phosphosulfate (PAPs). The SEM and micro-Raman spectroscopy results showed the hallmark feature of MCD. Further the gene expression studies elucidated the direct effect of sulfation inhibition on the WNT pathway, that in turn downregulated production of matrix metalloproteinases (MMPs), which causes abnormal matrix deposits leading to loss of transparency in vivo. It also resulted in downregulation of integrin and cadherin complexation that leads to disruption of the epithelial layer in the MCD affected corneas. This study offers a promising initial step toward establishing a relevant in vitro MCD disease model, to assess signaling transduction pathways and devise potential treatment strategies based on MMP administration to the MCD affected corneas.


Asunto(s)
Córnea/patología , Distrofias Hereditarias de la Córnea/patología , Células del Estroma/patología , Animales , Células Cultivadas , Cloratos/toxicidad , Córnea/efectos de los fármacos , Córnea/metabolismo , Córnea/ultraestructura , Distrofias Hereditarias de la Córnea/metabolismo , Inhibidores Enzimáticos/toxicidad , Epitelio Corneal/efectos de los fármacos , Epitelio Corneal/metabolismo , Epitelio Corneal/patología , Epitelio Corneal/ultraestructura , Proteínas del Ojo/antagonistas & inhibidores , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ontología de Genes , Cabras , Herbicidas/toxicidad , Procesamiento de Imagen Asistido por Computador , Metaloproteinasas de la Matriz/química , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Microscopía Electrónica de Rastreo , Microtecnología , Complejos Multienzimáticos/antagonistas & inhibidores , Complejos Multienzimáticos/metabolismo , Espectrometría Raman , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células del Estroma/ultraestructura , Sulfato Adenililtransferasa/antagonistas & inhibidores , Sulfato Adenililtransferasa/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
8.
Biomacromolecules ; 16(12): 3936-44, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26575529

RESUMEN

Controlling the mechanism of self-assembly in proteins has emerged as a potent tool for various biomedical applications. Silk fibroin self-assembly consists of gradual conformational transition from random coil to ß-sheet structure. In this work we elucidated the intermediate secondary conformation in the presence of Ca(2+) ions during fibroin self-assembly. The interaction of fibroin and calcium ions resulted in a predominantly α-helical intermediate conformation, which was maintained to certain extent even in the final conformation as illustrated by circular dichroism and attenuated total reflectance-Fourier transform infrared spectroscopy. Further, to elucidate the mechanism behind this interaction molecular modeling of the N-terminal region of fibroin with Ca(2+) ions was performed. Negatively charged glutamate and aspartate amino acids play a key role in the electrostatic interaction with positively charged calcium ions. Therefore, insights about modulation of self-assembly mechanism of fibroin could potentially be utilized to develop silk-based biomaterials consisting of the desired secondary conformation.


Asunto(s)
Ácido Aspártico/química , Bombyx/química , Calcio/química , Fibroínas/química , Ácido Glutámico/química , Animales , Bombyx/fisiología , Cationes Bivalentes , Dicroismo Circular , Fibroínas/aislamiento & purificación , Simulación de Dinámica Molecular , Estructura Secundaria de Proteína , Espectroscopía Infrarroja por Transformada de Fourier , Electricidad Estática
9.
J Mater Chem B ; 12(25): 6203-6220, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38833304

RESUMEN

RNA-based therapeutics have exhibited remarkable potential in targeting genetic factors for disease intervention, exemplified by recent mRNA vaccines for COVID-19. Nevertheless, the intrinsic instability of RNA and challenges related to its translational efficiency remain significant obstacles to the development of RNA as therapeutics. This study introduces an innovative RNA delivery approach using a silk fibroin (SF) and positively charged gelatin (Gel) hydrogel matrix to enhance RNA stability for controlled release. As a proof of concept, whole-cell RNA was incorporated into the hydrogel to enhance interactions with RNA molecules. Additionally, molecular modeling studies were conducted to explore the interactions between SF, collagen, chitosan (Chi), and the various RNA species including ribosomal RNAs (28S, 18S, 8.5S, and 5S rRNAs), transfer RNAs (tRNA-ALA, tRNA-GLN, and tRNA-Leu), as well as messenger RNAs (mRNA-GAPDH, mRNA-ß actin, and mRNA-Nanog), shedding light on the RNA-polymer interaction and RNA stability; SF exhibits a more robust interaction with RNA compared to collagen/gel and chitosan. We confirmed the molecular interactions of SF and RNA by FTIR and Raman spectroscopy, which were further supported by AFM and contact angle measurement. This research introduces a novel RNA delivery platform and insights into biopolymer-RNA interactions, paving the way for tailored RNA delivery systems in therapeutics and biomedical applications.


Asunto(s)
Gelatina , Hidrogeles , Gelatina/química , Hidrogeles/química , Humanos , Fibroínas/química , Portadores de Fármacos/química , Seda/química , Quitosano/química , Animales , ARN Mensajero/química , ARN Mensajero/genética , ARN de Transferencia/química , ARN de Transferencia/genética , ARN/química , Estabilidad del ARN , COVID-19 , SARS-CoV-2/genética
10.
Adv Healthc Mater ; 13(10): e2303513, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38291832

RESUMEN

The transforming growth factor-ß class of cytokines plays a significant role in articular cartilage formation from mesenchymal condensation to chondrogenic differentiation. However, their exogenous addition to the chondrogenic media makes the protocol expensive. It reduces the bioavailability of the cytokine to the cells owing to their burst release. The present study demonstrates an advanced bioconjugation strategy to conjugate transforming growth factor-ß3 (TGFß3) with silk fibroin matrix covalently via a cyanuric chloride coupling reaction. The tethering and change in secondary conformation are confirmed using various spectroscopic analyses. To assess the functionality of the chemically modified silk matrix, human bone marrow-derived mesenchymal stem cells (hBMSCs) and chondrocytes are cultured for 28 days in a chondrogenic differentiation medium. Gene expression and histological analysis reveal enhanced expression of chondrogenic markers with intense Safranin-O and Alcian Blue staining in TGFß3 conjugated silk matrices than where TGFß3 is exogenously added to the media for both hBMSCs and chondrocytes. Therefore, this study successfully recapitulates the native niche of TGFß3 and the role of the silk as a growth factor stabilizer. When cultured over TGFß3 conjugated silk matrices, hBMSCs display increased proteoglycan secretion and maximum chondrogenic trait with attenuation of chondrocyte hypertrophy over human chondrocytes.


Asunto(s)
Cartílago Articular , Fibroínas , Humanos , Cartílago Articular/metabolismo , Diferenciación Celular , Condrocitos , Condrogénesis , Fibroínas/química , Seda/metabolismo , Ingeniería de Tejidos/métodos , Factor de Crecimiento Transformador beta3/farmacología , Factor de Crecimiento Transformador beta3/metabolismo , Factores de Crecimiento Transformadores/metabolismo
11.
J Mater Chem B ; 12(38): 9508-9523, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39225012

RESUMEN

Silk is a widely accepted biomaterial for tissue regeneration owing to its tunable biomechanical properties and ease of chemical modification. However, a number of aspects associated with its clinical use are still debated. Indeed, to achieve clinical success, a biomaterial must favorably interact with host tissues without evoking local or systemic immuno-inflammatory responses. The analysis of immune responses associated with silk under in vitro and in vivo conditions provides useful insights, improving the understanding of the functional characteristics of silk biomaterials and further promoting their clinical application. Silk evokes moderate immune responses upon implantation in vivo, depending on the material structure, fabrication method, degradation time, and implantation in soft or hard tissue sites, which rapidly subside within a few days/weeks. In vitro studies indicate that its immune-stimulatory properties are largely due to inherent protein conformation and differential processing parameters. Strategically controlled levels of immune responses in vivo with marginal immunogenicity of silk-based biomaterials may contribute to matrix remodeling and replacement by native tissue matrix around the implanted site. Therefore, immunomodulatory strategies should be developed to promote the use of silk-based biomaterials as promising candidates for numerous clinical applications.


Asunto(s)
Materiales Biocompatibles , Seda , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Seda/química , Animales , Humanos , Ensayo de Materiales
12.
ACS Appl Mater Interfaces ; 16(8): 9925-9943, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38362893

RESUMEN

Implantation of a phenotypically stable cartilage graft could represent a viable approach for repairing osteoarthritic (OA) cartilage lesions. In the present study, we investigated the effects of modulating the bone morphogenetic protein (BMP), transforming growth factor beta (TGFß), and interleukin-1 (IL-1) signaling cascades in human bone marrow stromal cell (hBMSC)-encapsulated silk fibroin gelatin (SF-G) bioink. The selected small molecules LDN193189, TGFß3, and IL1 receptor antagonist (IL1Ra) are covalently conjugated to SF-G biomaterial to ensure sustained release, increased bioavailability, and printability, confirmed by ATR-FTIR, release kinetics, and rheological analyses. The 3D bioprinted constructs with chondrogenically differentiated hBMSCs were incubated in an OA-inducing medium for 14 days and assessed through a detailed qPCR, immunofluorescence, and biochemical analyses. Despite substantial heterogeneity in the observations among the donors, the IL1Ra molecule illustrated the maximum efficiency in enhancing the expression of articular cartilage components, reducing the expression of hypertrophic markers (re-validated by the GeneMANIA tool), as well as reducing the production of inflammatory molecules by the hBMSCs. Therefore, this study demonstrated a novel strategy to develop a chemically decorated, printable and biomimetic SF-G bioink to produce hyaline cartilage grafts resistant to acquiring OA traits that can be used for the treatment of degenerated cartilage lesions.


Asunto(s)
Bioimpresión , Cartílago Articular , Fibroínas , Humanos , Fibroínas/química , Cartílago Articular/metabolismo , Materiales Biocompatibles/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Diferenciación Celular , Gelatina/farmacología , Gelatina/química , Andamios del Tejido/química , Ingeniería de Tejidos , Impresión Tridimensional
13.
Biomed Mater ; 19(2)2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38198731

RESUMEN

The molecular niche of an osteoarthritic microenvironment comprises the native chondrocytes, the circulatory immune cells, and their respective inflammatory mediators. Although M2 macrophages infiltrate the joint tissue during osteoarthritis (OA) to initiate cartilage repair, the mechanistic crosstalk that dwells underneath is still unknown. Our study established a co-culture system of human OA chondrocytes and M2 macrophages in 3D spheroids and 3D bioprinted silk-gelatin constructs. It is already well established that Silk fibroin-gelatin bioink supports chondrogenic differentiation due to upregulation of the Wnt/ß-catenin pathway. Additionally, the presence of anti-inflammatory M2 macrophages significantly upregulated the expression of chondrogenic biomarkers (COL-II, ACAN) with an attenuated expression of the chondrocyte hypertrophy (COL-X), chondrocyte dedifferentiation (COL-I) and matrix catabolism (MMP-1 and MMP-13) genes even in the absence of the interleukins. Furthermore, the 3D bioprinted co-culture model displayed an upper hand in stimulating cartilage regeneration and OA inhibition than the spheroid model, underlining the role of silk fibroin-gelatin in encouraging chondrogenesis. Additionally, the 3D bioprinted silk-gelatin constructs further supported the maintenance of stable anti-inflammatory phenotype of M2 macrophage. Thus, the direct interaction between the primary OAC and M2 macrophages in the 3D context, along with the release of the soluble anti-inflammatory factors by the M2 cells, significantly contributed to a better understanding of the molecular mechanisms responsible for immune cell-mediated OA healing.


Asunto(s)
Bioimpresión , Fibroínas , Osteoartritis , Humanos , Condrocitos , Gelatina , Macrófagos/metabolismo , Antiinflamatorios
14.
Adv Healthc Mater ; : e2402024, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39226530

RESUMEN

In the pursuit of new wound care products, researchers are exploring methods to improve wound healing through exogenous wound healing products. However, diverging from this conventional approach, this work has developed an endogenous support system for wound healing, drawing inspiration from the body's innate healing mechanisms governed by the sequential release of metal ions by body at wound site to promote different stages of wound healing. This work engineers a multi-ion-releasing sprayable hydrogel system, to mimic this intricate process, representing the next evolutionary step in wound care products. It comprises Alginate (Alg) and Fibrin (Fib) hydrogel infused with Polylactic acid (PLA) polymeric microcarriers encapsulating multi (calcium, copper, and zinc) nanoparticles (Alg-Fib-PLA-nCMB). Developed sprayable Alg-Fib-PLA-nCMB hydrogel show sustained release of beneficial multi metallic ions at wound site, offering a range of advantages including enhanced cellular function, antibacterial properties, and promotion of crucial wound healing processes like cell migration, ROS mitigation, macrophage polarization, collagen deposition, and vascular regeneration. In a comparative study with a commercial product (Midstress spray), developed Alg-Fib-PLA-nCMB hydrogel demonstrates superior wound healing outcomes in a rat model, indicating its potential for next generation wound care product, addressing critical challenges and offering a promising avenue for future advancements in the wound management.

15.
Biofabrication ; 16(3)2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38507802

RESUMEN

3D bioprinting has the potential for the rapid and precise engineering of hydrogel constructs that can mimic the structural and optical complexity of a healthy cornea. However, the use of existing light-activated bioinks for corneal printing is limited by their poor cytocompatibility, use of cytotoxic photoinitiators (PIs), low photo-crosslinking efficiency, and opaque/colored surface of the printed material. Herein, we report a fast-curable, non-cytotoxic, optically transparent bioprinting system using a new water-soluble benzoyl phosphinate-based PI and photocrosslinkable methacrylated hyaluronic acid (HAMA). Compared with commercially available PIs, the newly developed PI, lithium benzoyl (phenyl) phosphinate (BP), demonstrated increased photoinitiation efficiency under visible light and low cytotoxicity. Using a catalytic amount of BP, the HA-based bioinks quickly formed 3D hydrogel constructs under low-energy visible-light irradiation (405 nm, <1 J cm-2). The mechanical properties and printability of photocurable bioinks were further improved by blending low (10 kDa) and high (100 kDa) molecular weight (MW) HAMA by forming multilength networks. For potential applications as corneal scaffolds, stromal cell-laden dome-shaped constructs were fabricated using MW-blended HAMA/BP bioink and a digital light processing printer. The HA-based photocurable bioinks exhibited good cytocompatibility (80%-95%), fast curing kinetics (<5 s), and excellent optical transparency (>90% in the visible range), potentially making them suitable for corneal tissue engineering.


Asunto(s)
Bioimpresión , Andamios del Tejido , Andamios del Tejido/química , Impresión Tridimensional , Ingeniería de Tejidos , Córnea , Hidrogeles , Células del Estroma , Luz
17.
Biomacromolecules ; 14(2): 311-21, 2013 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-23305127

RESUMEN

Direct-write assembly allows rapid fabrication of complex three-dimensional (3D) architectures, such as scaffolds simulating anatomical shapes, avoiding the need for expensive lithographic masks. However, proper selection of polymeric ink composition and tailor-made viscoelastic properties are critically important for smooth deposition of ink and shape retention. Deposition of only silk solution leads to frequent clogging due to shear-induced ß-sheet crystallization, whereas optimized viscoelastic property of silk-gelatin blends facilitate the flow of these blends through microcapillary nozzles of varying diameter. This study demonstrates that induction of controlled changes in scaffold surface chemistry, by optimizing silk-gelatin ratio, can govern cell proliferation and maintenance of chondrocyte morphology. Microperiodic silk-gelatin scaffolds can influence postexpansion redifferentiation of goat chondrocytes by enhancing Sox-9 gene expression, aggregation, and driving cartilage matrix production, as evidenced by upregulation of collagen type II and aggrecan expression. The strategy for optimizing redifferentiation of chondrocytes can offer valuable consideration in scaffold-based cartilage repair strategies.


Asunto(s)
Condrocitos/metabolismo , Gelatina/metabolismo , Seda/metabolismo , Ingeniería de Tejidos , Andamios del Tejido , Agrecanos/biosíntesis , Agrecanos/metabolismo , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/metabolismo , Bombyx , Cartílago/citología , Cartílago/crecimiento & desarrollo , Adhesión Celular , Técnicas de Cultivo de Célula , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Colágeno Tipo II/biosíntesis , Gelatina/química , Cabras , Ensayo de Materiales , Factor de Transcripción SOX9/biosíntesis , Factor de Transcripción SOX9/metabolismo , Seda/química , Espectroscopía Infrarroja por Transformada de Fourier , Sustancias Viscoelásticas
18.
Biotechnol Adv ; 69: 108273, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37863444

RESUMEN

The long duration space missions across the Low Earth Orbit (LEO) often expose the voyagers to an abrupt zero gravity influence. The severe extraterrestrial cosmic radiation directly causes a plethora of moderate to chronic healthcare crises. The only feasible solution to manage critical injuries on board is surgical interventions or immediate return to Earth. This led the group of space medicine practitioners to adopt principles from tissue engineering and develop human tissue equivalents as an immediate regenerative therapy on board. The current review explicitly demonstrates the constructive application of different tissue-engineered equivalents matured under the available ground-based microgravity simulation facilities. Further, it elucidates how augmenting the superiority of biomaterial-based 3D bioprinting technology can enhance their clinical applicability. Additionally, the regulatory role of weightlessness condition on the underlying cellular signaling pathways governing tissue morphogenesis has been critically discussed. This information will provide future directions on how 3D biofabrication can be used as a plausible tool for healing on-flight chronic health emergencies. Thus, in our review, we aimed to precisely debate whether 3D biofabrication is deployed to cater to on-flight healthcare anomalies or space-like conditions are being utilized for generating 3D bioprinted human tissue constructs for efficient drug screening and regenerative therapy.


Asunto(s)
Bioimpresión , Ingeniería de Tejidos , Humanos , Materiales Biocompatibles , Cicatrización de Heridas , Andamios del Tejido
19.
Biomed Mater ; 18(6)2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37860885

RESUMEN

Stem cell-based tissue engineering is an emerging tool for developing functional tissues of choice. To understand pluripotency and hepatic differentiation of mouse embryonic stem cells (mESCs) on a three-dimensional (3D) scaffold, we established an efficient approach for generating hepatocyte-like cells (HLCs) from hepatoblast cells. We developed porous and biodegradable scaffold, which was stimulated with exogenous growth factors and investigated stemness and differentiation capacity of mESCs into HLCs on the scaffoldin-vitro. In animal studies, we had cultured mESCs-derived hepatoblast-like cells on the scaffold and then, transplanted them into the partially hepatectomized C57BL/6 male mice model to evaluate the effect of gelatin scaffold on hepatic regeneration. The 3D culture system allowed maintenance of stemness properties in mESCs. The step-wise induction of mESCs with differentiation factors leads to the formation of HLCs and expressed liver-specific genes, including albumin, hepatocyte nucleic factor 4 alpha, and cytokeratin 18. In addition, cells also expressed Ki67, indicating cells are proliferating. The secretome showed expression of albumin, urea, creatinine, alanine transaminase, and aspartate aminotransferase. However, the volume of the excised liver which aids regeneration has not been studied. Our results indicate that hepatoblast cells on the scaffold implanted in PH mouse indicates that these cells efficiently differentiate into HLCs and cholangiocytes, forming hepatic lobules with central and portal veins, and bile duct-like structures with neovascularization. The gelatin scaffold provides an efficient microenvironment for liver differentiation and regeneration bothin-vitroandin-vivo. These hepatoblasts cells would be a valuable source for 3D liver tissue engineering/transplantation in liver diseases.


Asunto(s)
Gelatina , Regeneración Hepática , Masculino , Animales , Ratones , Gelatina/química , Ratones Endogámicos C57BL , Hígado/cirugía , Diferenciación Celular , Hepatocitos , Proliferación Celular , Albúminas
20.
Tissue Eng Part A ; 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37950715

RESUMEN

Manual grading of cartilage histology images for investigating the extent and severity of osteoarthritis (OA) involves critical examination of the cell characteristics, which makes this task tiresome, tedious, and error prone. This results in wide interobserver variation, causing ambiguities in OA grade prediction. Such drawbacks of manual assessment can be overcome by implementing artificial intelligence-based automated image classification techniques such as deep learning (DL). Hence, we present the feasibility of training a deep neural network with cartilage histology images, which can grade the extent and severity of knee OA based on modified Mankin scoring system. The grading system used here for automating OA grading was simplified and modified based on the microscopic observations from the histology images, where three parameters (Safranin-O staining intensity, chondrocyte distribution and arrangement, and morphology) were considered for evaluating the OA progression. The histology images were tiled, labeled, and grouped together based on the developed grading system (Grade 0-3). Four different DL architectures were tried for image classification and the best performing model was selected by fivefold validation method. With a validation accuracy of ∼84%, 0.632 Cohen's kappa score, and an excellent receiver operating characteristic (ROC)-area under the ROC curve ranging between 0.89 and 0.99, DenseNet121 was selected among the four models as the best performing model, and was used for inferencing on new data. Final grades obtained from the models were in accordance with the grades provided by the medical experts. We hereby demonstrate that a DL architecture can be taught to interpret the degree of cartilage degradation, with excellent discriminatory ability across all four classes of OA severity. Unlike other works where radiographic images have been considered for grading of OA, we have considered histology images, which is a fundamental approach for grading OA extent and severity. This would bring a paradigm shift in histology-based assessment of OA, making this automated approach to be explored as an option for OA grading standardization. Ethical approval number-IAH-BMR-018/10-19.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA