Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Nanobiotechnology ; 21(1): 310, 2023 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-37658367

RESUMO

BACKGROUND: Basic fibroblast growth factor (bFGF) is one of the critical components accelerating angiogenesis and tissue regeneration by promoting the migration of dermal fibroblasts and endothelial cells associated with matrix formation and remodeling in wound healing process. However, clinical applications of bFGF are substantially limited by its unstable nature due to rapid decomposition under physiological microenvironment. RESULTS: In this study, we present the bFGF-loaded human serum albumin nanoparticles (HSA-bFGF NPs) as a means of enhanced stability and sustained release platform during tissue regeneration. Spherical shape of the HSA-bFGF NPs with uniform size distribution (polydispersity index < 0.2) is obtained via a simple desolvation and crosslinking process. The HSA-bFGF NPs securely load and release the intact soluble bFGF proteins, thereby significantly enhancing the proliferation and migration activity of human dermal fibroblasts. Myofibroblast-related genes and proteins were also significantly down-regulated, indicating decrease in risk of scar formation. Furthermore, wound healing is accelerated while achieving a highly organized extracellular matrix and enhanced angiogenesis in vivo. CONCLUSION: Consequently, the HSA-bFGF NPs are suggested not only as a delivery vehicle but also as a protein stabilizer for effective wound healing and tissue regeneration.


Assuntos
Fator 2 de Crescimento de Fibroblastos , Nanopartículas , Humanos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Células Endoteliais , Albumina Sérica Humana , Cicatrização
2.
Small ; 18(22): e2200757, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35521748

RESUMO

Numerous methods have been introduced to produce 3D cell cultures that can reduce the need for animal experimentation. This study presents a unique 3D culture platform that features bioinspired strands of electrospun nanofibers (BSeNs) and aquatic cell lines to compensate for shortcomings in the current cell spheroid generation techniques. The use of BSeNs in 3D zebrafish liver cell cultures is found to improve liver and reproductive functions through spheroid-based in vitro assays such as whole transcriptome sequencing and reproductive toxicity testing, with optimized properties exhibiting results similar to those obtained for fish embryo acute toxicity (FET, OECD TG 236) following exposure to environmental endocrine-disrupting chemicals (17ß-Estradiol (E2), 4-hydroxytamoxifen (4-HT), and bisphenol compounds (bisphenol A (BPA) and 9,9-Bis(4-hydroxyphenyl)fluorene (BPFL)). These findings indicate that the beneficial effects of bioinspired materials that closely mimic ECM environments can yield efficient zebrafish cells with intrinsic functions and xenobiotic metabolism similar to those of zebrafish embryos. As a closer analog for the in vivo conditions that are associated with exposure to potentially hazardous chemicals, the straightforward culture model introduced in this study shows promise as an alternative tool that can be used to further eco-environmental assessment.


Assuntos
Disruptores Endócrinos , Peixe-Zebra , Animais , Disruptores Endócrinos/metabolismo , Disruptores Endócrinos/toxicidade , Fígado/metabolismo , Esferoides Celulares/metabolismo , Testes de Toxicidade , Peixe-Zebra/metabolismo
3.
Small ; 12(45): 6266-6278, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27717233

RESUMO

Using small interfering RNA (siRNA) to regulate gene expression is an emerging strategy for stem cell manipulation to improve stem cell therapy. However, conventional methods of siRNA delivery into stem cells based on solution-mediated transfection are limited due to low transfection efficiency and insufficient duration of cell-siRNA contact during lengthy culturing protocols. To overcome these limitations, a bio-inspired polymer-mediated reverse transfection system is developed consisting of implantable poly(lactic-co-glycolic acid) (PLGA) scaffolds functionalized with siRNA-lipidoid nanoparticle (sLNP) complexes via polydopamine (pDA) coating. Immobilized sLNP complexes are stably maintained without any loss of siRNA on the pDA-coated scaffolds for 2 weeks, likely due to the formation of strong covalent bonds between amine groups of sLNP and catechol group of pDA. siRNA reverse transfection with the pDA-sLNP-PLGA system does not exhibit cytotoxicity and induces efficient silencing of an osteogenesis inhibitor gene in human adipose-derived stem cells (hADSCs), resulting in enhanced osteogenic differentiation of hADSCs. Finally, hADSCs osteogenically committed on the pDA-sLNP-PLGA scaffolds enhanced bone formation in a mouse model of critical-sized bone defect. Therefore, the bio-inspired reverse transfection system can provide an all-in-one platform for genetic modification, differentiation, and transplantation of stem cells, simultaneously enabling both stem cell manipulation and tissue engineering.


Assuntos
Tecido Adiposo/citologia , Diferenciação Celular/fisiologia , Osteogênese/fisiologia , Células-Tronco/citologia , Regeneração Óssea/genética , Regeneração Óssea/fisiologia , Diferenciação Celular/genética , Humanos , Ácido Láctico/química , Osteogênese/genética , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , RNA Interferente Pequeno/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química
4.
Small ; 11(17): 2069-79, 2015 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-25515928

RESUMO

Stem cells are poorly permissive to non-viral gene transfection reagents. In this study, we explored the possibility of improving gene delivery into human embryonic (hESC) and mesenchymal (hMSC) stem cells by synergizing the activity of a cell-binding ligand with a polymer that releases nucleic acids in a cytoplasm-responsive manner. A 29 amino acid long peptide, RVG, targeting the nicotinic acetylcholine receptor (nAchR) was identified to bind both hMSC and H9-derived hESC. Conjugating RVG to a redox-sensitive biodegradable dendrimer-type arginine-grafted polymer (PAM-ABP) enabled nanoparticle formation with plasmid DNA without altering the environment-sensitive DNA release property and favorable toxicity profile of the parent polymer. Importantly, RVG-PAM-ABP quantitatively enhanced transfection into both hMSC and hESC compared to commercial transfection reagents like Lipofectamine 2000 and Fugene. ∼60% and 50% of hMSC and hESC were respectively transfected, and at increased levels on a per cell basis, without affecting pluripotency marker expression. RVG-PAM-ABP is thus a novel bioreducible, biocompatible, non-toxic, synthetic gene delivery system for nAchR-expressing stem cells. Our data also demonstrates that a cell-binding ligand like RVG can cooperate with a gene delivery system like PAM-ABP to enable transfection of poorly-permissive cells.


Assuntos
Materiais Biocompatíveis/química , Técnicas de Transferência de Genes , Vetores Genéticos/química , Peptídeos/química , Células-Tronco/citologia , Animais , Arginina/química , Citoplasma/metabolismo , DNA/química , Células-Tronco Embrionárias/citologia , Fibroblastos/metabolismo , Citometria de Fluxo , Proteínas de Fluorescência Verde/química , Humanos , Ligantes , Lipídeos/química , Células-Tronco Mesenquimais/citologia , Camundongos , Oxirredução , Fenótipo , Plasmídeos/metabolismo , Polímeros/química , Receptores Nicotínicos/metabolismo , Transfecção
5.
Biomacromolecules ; 16(9): 2541-55, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26280621

RESUMO

Current advances in biomaterial fabrication techniques have broadened their application in different realms of biomedical engineering, spanning from drug delivery to tissue engineering. The success of biomaterials depends highly on the ability to modulate cell and tissue responses, including cell adhesion, as well as induction of repair and immune processes. Thus, most recent approaches in the field have concentrated on functionalizing biomaterials with different biomolecules intended to evoke cell- and tissue-specific reactions. Marine mussels produce mussel adhesive proteins (MAPs), which help them strongly attach to different surfaces, even under wet conditions in the ocean. Inspired by mussel adhesiveness, scientists discovered that dopamine undergoes self-polymerization at alkaline conditions. This reaction provides a universal coating for metals, polymers, and ceramics, regardless of their chemical and physical properties. Furthermore, this polymerized layer is enriched with catechol groups that enable immobilization of primary amine or thiol-based biomolecules via a simple dipping process. Herein, this review explores the versatile surface modification techniques that have recently been exploited in tissue engineering and summarizes polydopamine polymerization mechanisms, coating process parameters, and effects on substrate properties. A brief discussion of polydopamine-based reactions in the context of engineering various tissue types, including bone, blood vessels, cartilage, nerves, and muscle, is also provided.


Assuntos
Bivalves/química , Materiais Revestidos Biocompatíveis/química , Dopamina/química , Indóis/química , Polímeros/química , Proteínas/química , Engenharia Tecidual/métodos , Animais , Humanos , Camundongos , Células NIH 3T3 , Propriedades de Superfície
6.
Biomacromolecules ; 15(1): 361-72, 2014 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-24304175

RESUMO

Peripheral arterial disease is a common manifestation of systemic atherosclerosis, which results in more serious consequences of ischemic events in peripheral tissues such as the lower extremities. Cell therapy has been tested as a treatment for peripheral ischemia that functions by inducing angiogenesis in the ischemic region. However, the poor survival and engraftment of transplanted cells limit the efficacy of cell therapy. In order to overcome such challenges, we applied genetically engineered cell sheets using a cell-interactive and thermosensitive hydrogel and nonviral polymer nanoparticles. C2C12 myoblast sheets were formed on Tetronic-tyramine (Tet-TA)-RGD hydrogel prepared through a highly efficient and noncytotoxic enzymatic reaction. The myoblast sheets were then transfected with vascular endothelial growth factor (VEGF) plasmids using poly(ß-amino ester) nanoparticles to increase the angiogenic potential of the sheets. The transfection increased the VEGF expression and secretion from the C2C12 sheets. The enhanced angiogenic effect of the VEGF-transfected C2C12 sheets was confirmed using an in vitro capillary formation assay. More importantly, the transplantation of the VEGF-transfected C2C12 sheets promoted the formation of capillaries and arterioles in ischemic muscles, attenuated the muscle necrosis and fibrosis progressed by ischemia, and eventually prevented ischemic limb loss. In conclusion, the combination of cell sheet engineering and genetic modification can provide more effective treatment for therapeutic angiogenesis.


Assuntos
Engenharia Genética/métodos , Mioblastos/fisiologia , Mioblastos/transplante , Neovascularização Patológica/genética , Neovascularização Patológica/cirurgia , Animais , Sobrevivência Celular/fisiologia , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/fisiologia , Células Endoteliais da Veia Umbilical Humana/transplante , Humanos , Isquemia/genética , Isquemia/cirurgia , Camundongos
7.
Biofabrication ; 16(2)2024 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-38198701

RESUMO

Advancements in biofabrication have led to major strides toward creating authentic organ models; however, replicating intricate organ structures without scaffolds remains challenging. In this study, we introduce a method utilizing surface-modifiable magnetic nanofibers to achieve precise control over spheroid functions and geometrical features, allowing the creation of multiple functional domains within a single microtissue. We generated magnetized nanofibers by electrospinning magnetic nanoparticles dispersed in poly-L-lactic acid solution. These fibers were then coated with polydopamine (PD) to enhance their biological functions, particularly reactive oxygen species (ROS) scavenging. These PD-coated magnetic fibers (PMFs) had magnetic-responsive properties when incorporated into human dermal fibroblast spheroids (0.019 ± 0.001 emu g-1). Furthermore, PMFs within the spheroids effectively regulated ROS levels by upregulating the expression of key anti-oxidative genes such assuperoxide dismutase-1(2.2 ± 0.1) andglutathione peroxidase-1(2.6 ± 0.1). By exploiting the magnetic responsiveness of spheroids, we were able to assemble them into various structures such as linear, triangular, and square structures using remotely applied magnetic forces. Within the assembled three-dimensional constructs, the cells in spheroids incorporating PMFs demonstrated resistance to ROS regulatory activity in the presence of hydrogen peroxide, while spheroids composed of bare fibers exhibited high ROS levels. Furthermore, we assembled spheroids containing fibroblasts and endothelial cells into complex tissue structures resembling vessels under magnetic manipulation. This innovative method holds tremendous promise for organ modeling and regenerative medicine due to the unprecedented control it allows in developing microtissues that closely emulate real organs.


Assuntos
Nanofibras , Esferoides Celulares , Humanos , Células Endoteliais , Nanofibras/química , Espécies Reativas de Oxigênio , Magnetismo , Engenharia Tecidual/métodos
8.
Adv Healthc Mater ; : e2400232, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38696729

RESUMO

Inorganic nanoparticles are promising materials for bone tissue engineering due to their chemical resemblance to the native bone structure. However, most studies are unable to capture the entirety of the defective environment, providing limited bone regenerative abilities. Hence, this study aims to develop a multifunctional nanoparticle to collectively control the defective bone niche, including immune, angiogenic, and osteogenic systems. The nanoparticles, self-assembled by biomimetic mineralization and tannic acid (TA)-mediated metal-polyphenol network (MPN), are released sustainably after the incorporation within a gelatin cryogel. The released nanoparticles display a reduction in M1 macrophages by means of reactive oxygen species (ROS) elimination. Consequently, osteoclast maturation is also reduced, which is observed by the minimal formation of multinucleated cells (0.4%). Furthermore, the proportion of M2 macrophages, osteogenic differentiation, and angiogenic potential are consistently increased by the effects of magnesium from the nanoparticles. This orchestrated control of multiple systems influences the in vivo vascularized bone regeneration in which 80% of the critical-sized bone defect is regenerated with new bones with mature lamellar structure and arteriole-scale micro-vessels. Altogether, this study emphasizes the importance of the coordinated modulation of immune, osteogenic, and angiogenic systems at the bone defect site for robust bone regeneration.

9.
Biofabrication ; 16(3)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38739412

RESUMO

Reconstruction of large 3D tissues based on assembly of micro-sized multi-cellular spheroids has gained attention in tissue engineering. However, formation of 3D adipose tissue from spheroids has been challenging due to the limited adhesion capability and restricted cell mobility of adipocytes in culture media. In this study, we addressed this problem by developing adipo-inductive nanofibers enabling dual delivery of indomethacin and insulin. These nanofibers were introduced into composite spheroids comprising human adipose-derived stem cells (hADSCs). This approach led to a significant enhancement in the formation of uniform lipid droplets, as evidenced by the significantly increased Oil red O-stained area in spheroids incorporating indomethacin and insulin dual delivery nanofibers (56.9 ± 4.6%) compared to the control (15.6 ± 3.5%) with significantly greater gene expression associated with adipogenesis (C/EBPA, PPARG, FABP4, and adiponectin) of hADSCs. Furthermore, we investigated the influence of culture media on the migration and merging of spheroids and observed significant decrease in migration and merging of spheroids in adipogenic differentiation media. Conversely, the presence of adipo-inductive nanofibers promoted spheroid fusion, allowing the formation of macroscopic 3D adipose tissue in the absence of adipogenic supplements while facilitating homogeneous adipogenesis of hADSCs. The approach described here holds promise for the generation of 3D adipose tissue constructs by scaffold-free assembly of stem cell spheroids with potential applications in clinical and organ models.


Assuntos
Adipogenia , Tecido Adiposo , Nanofibras , Esferoides Celulares , Células-Tronco , Engenharia Tecidual , Nanofibras/química , Humanos , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Insulina/metabolismo , Indometacina/farmacologia , Adipócitos/citologia , Adipócitos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Alicerces Teciduais/química , Adiponectina/metabolismo , Células Cultivadas
10.
Biofabrication ; 16(2)2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38447223

RESUMO

Recent advances in regenerative medicine and tissue engineering have enabled the biofabrication of three-dimensional (3D) tissue analogues with the potential for use in transplants and disease modeling. However, the practical use of these biomimetic tissues has been hindered by the challenge posed by reconstructing anatomical-scale micro-vasculature tissues. In this study, we suggest that co-cultured spheroids within hydrogels hold promise for regenerating highly vascularized and innervated tissues, bothin vitroandin vivo. Human adipose-derived stem cells (hADSCs) and human umbilical vein cells (HUVECs) were prepared as spheroids, which were encapsulated in gelatin methacryloyl hydrogels to fabricate a 3D pre-vascularized tissue. The vasculogenic responses, extracellular matrix production, and remodeling depending on parameters like co-culture ratio, hydrogel strength, and pre-vascularization time forin vivointegration with native vessels were then delicately characterized. The co-cultured spheroids with 3:1 ratio (hADSCs/HUVECs) within the hydrogel and with a pliable storage modulus showed the greatest vasculogenic potential, and ultimately formedin vitroarteriole-scale vasculature with a longitudinal lumen structure and a complex vascular network after long-term culturing. Importantly, the pre-vascularized tissue also showed anastomotic vascular integration with host blood vessels after transplantation, and successful vascularization that was positive for both CD31 and alpha-smooth muscle actin covering 18.6 ± 3.6µm2of the luminal area. The described co-cultured spheroids-laden hydrogel can therefore serve as effective platform for engineering 3D vascularized complex tissues.


Assuntos
Hidrogéis , Engenharia Tecidual , Humanos , Engenharia Tecidual/métodos , Hidrogéis/química , Técnicas de Cocultura , Células Endoteliais da Veia Umbilical Humana , Medicina Regenerativa , Alicerces Teciduais/química
11.
Tissue Eng Part A ; 30(5-6): 225-243, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38062771

RESUMO

A combination of hydrogels and stem cell spheroids has been used to engineer three-dimensional (3D) osteochondral tissue, but precise zonal control directing cell fate within the hydrogel remains a challenge. In this study, we developed a composite spheroid-laden bilayer hydrogel to imitate osteochondral tissue by spatially controlled differentiation of human adipose-derived stem cells. Meticulous optimization of the spheroid-size and mechanical strength of gelatin methacryloyl (GelMA) hydrogel enables the cells to homogeneously sprout within the hydrogel. Moreover, fibers immobilizing transforming growth factor beta-1 (TGF-ß1) or bone morphogenetic protein-2 (BMP-2) were incorporated within the spheroids, which induced chondrogenic or osteogenic differentiation of cells in general media, respectively. The spheroids-filled GelMA solution was crosslinked to create the bilayer hydrogel, which demonstrated a strong interfacial adhesion between the two layers. The cell sprouting enhanced the adhesion of each hydrogel, demonstrated by increase in tensile strength from 4.8 ± 0.4 to 6.9 ± 1.2 MPa after 14 days of culture. Importantly, the spatially confined delivery of BMP-2 within the spheroids increased mineral deposition and more than threefold enhanced osteogenic genes of cells in the bone layer while the cells induced by TGF-ß1 signals were apparently differentiated into chondrocytes within the cartilage layer. The results suggest that our composite spheroid-laden hydrogel could be used for the biofabrication of osteochondral tissue, which can be applied to engineer other complex tissues by delivery of appropriate biomolecules.


Assuntos
Osteogênese , Fator de Crescimento Transformador beta1 , Humanos , Fator de Crescimento Transformador beta1/farmacologia , Hidrogéis/farmacologia , Engenharia Tecidual/métodos , Diferenciação Celular , Alicerces Teciduais
12.
Bioact Mater ; 36: 185-202, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38463552

RESUMO

Wound healing in cases of excessive inflammation poses a significant challenge due to compromised neovascularization. Here, we propose a multi-functional composite hydrogel engineered to overcome such conditions through recruitment and activation of macrophages with adapted degradation of the hydrogel. The composite hydrogel (G-TSrP) is created by combining gelatin methacryloyl (GelMA) and nanoparticles (TSrP) composed of tannic acid (TA) and Sr2+. These nanoparticles are prepared using a one-step mineralization process assisted by metal-phenolic network formation. G-TSrP exhibits the ability to eliminate reactive oxygen species and direct polarization of macrophages toward M2 phenotype. It has been observed that the liberation of TA and Sr2+ from G-TSrP actively facilitate the recruitment and up-regulation of the expression of extracellular matrix remodeling genes of macrophages, and thereby, coordinate in vivo adapted degradation of the G-TSrP. Most significantly, G-TSrP accelerates angiogenesis despite the TA's inhibitory properties, which are counteracted by the released Sr2+. Moreover, G-TSrP enhances wound closure under inflammation and promotes normal tissue formation with strong vessel growth. Genetic analysis confirms macrophage-mediated wound healing by the composite hydrogel. Collectively, these findings pave the way for the development of biomaterials that promote wound healing by creating regenerative environment.

13.
Biochem Biophys Res Commun ; 430(2): 793-7, 2013 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-23206696

RESUMO

Recently, it was reported that bone morphogenetic protein 4 (BMP4) alone or BMP4 combined with fibroblast growth factor 2 (FGF2) treatment enhanced mesodermal differentiation of human embryonic stem cells (hESCs) that were cultured feeder-free on Matrigel. In this study, we show that mesodermal lineage-induced embryoid bodies (EBs) generate greater numbers of osteogenic and chondrogenic lineage cells. To induce the mesodermal lineage, hESCs were treated with BMP4 and FGF2 during the EB state. Quantitative real-time reverse transcription-polymerase chain reaction analysis showed that the treatment decreased endodermal and ectodermal lineage gene expression and increased mesodermal lineage gene expression. Importantly, the mesodermal lineage-induced EBs underwent enhanced osteogenic and chondrogenic differentiation after differentiation induction. This method could be useful to enhance the osteogenic or chondrogenic differentiation of hESCs.


Assuntos
Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Mesoderma/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Células Cultivadas , Corpos Embrioides/efeitos dos fármacos , Humanos , Mesoderma/citologia
14.
Biomacromolecules ; 14(12): 4309-19, 2013 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-24279345

RESUMO

In this study, thermosensitive hydrogels incorporated with multiple cell-interactive factors were developed as a substrate to form monolayer of human umbilical vein endothelial cells (HUVECs) that can be detached and transferrable to target sites as a cell-sheet in response to temperature change. The cell adhesive peptide (RGD) and growth factor (bFGF) covalently incorporated within the hydrogel significantly enhanced adhesion and proliferation of HUVECs, allowing for the formation of their confluent monolayer. Meanwhile, the precisely controllable change in the size of the hydrogels was observed by a repeated increase and decrease in temperature from 37 to 4 °C. By exploiting this unique behavior, the detachment and transfer of HUVEC sheet confluently cultured at 37 °C was rapidly induced within 10 min by expansion of the hydrogels when the temperature was decreased to 4 °C. The transferred cell sheet was highly viable and maintained robust cell-cell junction. Finally, the process of cell sheet transfer was directly applied onto an ischemic injury in the hind limb of mice. The transplanted HUVECs as a sheet retarded tissue necrosis over 14 days in comparison with that of direct injection of the same number of cells. Our results suggest that the developed multifunctional Tetronic-tyramine hydrogels could serve as an ideal substrate to modulate the formation of an endothelial cell layer that could potentially be utilized to treat peripheral arterial disease.


Assuntos
Células Endoteliais da Veia Umbilical Humana/fisiologia , Hidrogéis/química , Isquemia/terapia , Neovascularização Fisiológica , Animais , Adesão Celular , Técnicas de Cultura de Células , Proliferação de Células , Forma Celular , Células Cultivadas , Feminino , Fator 2 de Crescimento de Fibroblastos/química , Membro Posterior/irrigação sanguínea , Células Endoteliais da Veia Umbilical Humana/transplante , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oligopeptídeos/química , Engenharia Tecidual
15.
Ann Plast Surg ; 70(1): 98-102, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22274147

RESUMO

Regeneration of volume-stable adipose tissue is required for treatment of soft-tissue loss due to cancer, trauma, burns and for correctional cosmetic surgery. In this study, we hypothesized that transplantation of human adipose-derived stromal cells (hADSCs) using polycaprolactone (PCL) scaffolds fabricated with a solid free-form fabrication method would better maintain the volume of regenerated adipose tissues, as compared with the use of fibrin gel. Six weeks after implantation into the dorsal subcutaneous pockets of athymic mice, the volumes and adipose tissue areas of hADSC-PCL scaffold implants were significantly larger than those of hADSC-fibrin implants. In addition, the mRNA expression of adipogenic genes was more extensive in the hADSC-PCL scaffold implants.


Assuntos
Regeneração Tecidual Guiada/métodos , Lipogênese , Poliésteres , Células Estromais/transplante , Gordura Subcutânea/fisiologia , Alicerces Teciduais , Adipogenia/genética , Animais , Células Cultivadas , Feminino , Fibrina , Marcadores Genéticos , Humanos , Lipogênese/genética , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Varredura , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Gordura Subcutânea/citologia
16.
Tissue Eng Part C Methods ; 29(10): 447-458, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37440328

RESUMO

Muscle tissue engineering has been the focus of extensive research due to its potential for numerous medical applications, including ex vivo actuator development and clinical treatments. In this study, we developed a method for harvesting muscle fiber in a floatable and translocatable manner utilizing thermally expandable hydrogels with a chemically patterned polydopamine (PD) layer generated by microcontact printing (µCP). The µCP of PD on the hydrogel facilitated the formation of stripe patterns with varying widths of printed/nonprinted area (50/50, 100/100, and 200/200 µm). The spatially controlled adhesion of C2C12 myoblasts on the PD patterns produced clearly distinguishable muscle fibers, and translocated muscle fibers exhibited preserved extracellular matrix and junction proteins. Furthermore, the development of anisotropic arrangements and mature myotubes within the fibers suggests the potential for functional control of engineered muscle tissues. Overall, the muscle fiber harvesting method developed herein is suitable for both translocation and floating and is a promising technique for muscle tissue engineering as it mimics the structure-function relationship of natural tissue.


Assuntos
Hidrogéis , Fibras Musculares Esqueléticas , Engenharia Tecidual/métodos , Mioblastos , Matriz Extracelular , Alicerces Teciduais
17.
Biomater Res ; 27(1): 132, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-38087321

RESUMO

BACKGROUND: Bone tissue regeneration is regulated by complex events, including inflammation, osteoinduction, and remodeling. Therefore, to induce the complete restoration of defective bone tissue, biomaterials with the ability to regulate the collective bone regenerative system are beneficial. Although some studies conclude that reducing reactive oxygen species created a favorable environment for bone regeneration by controlling inflammation, biomaterials that can simultaneously promote osteogenesis and regulate inflammation have not been developed. Herein, we describe the development of a multi-functional nanoparticle and its hydrogel composite with osteoinductive, anti-inflammatory, and osteoclast-maturation regulatory functions for enhanced bone regeneration. METHODS: Tannic acid-mineral nanoparticles (TMP) were prepared by self-assembly of tannic acid in an ion-rich simulated body fluid containing Ca2+ and PO43-. Particles with a diameter of 443 ± 91 nm were selected for their stable spherical morphology and minimal tendency to aggregate. The particles were homogeneously embedded within a gelatin-based cryogel (TMP/Gel) to be used in further experiments. The osteoinductive properties, anti-inflammatory and osteoclast-maturation regulatory functions in vitro were tested by culturing corresponding cells on either TMP/Gel or a gelatin-based cryogel without the particles (Gel). For in vivo analyses, a murine calvarial defect model was used. Statistical analyses were carried out using a Graphpad Prism 7 software (San Diego, CA, USA) to perform one-way analysis of variance ANOVA with Tukey's honest significant difference test and a Student's t-test (for two variables) (P < 0.05). RESULTS: Excellent biocompatibility and radical scavenging abilities were exhibited by the TMP/Gel. The expression of osteogenic mRNA is significantly increased in human adipose-derived stem cells seeded on the TMP/Gel compared to those without the particles. Furthermore, RAW264.7 cells seeded on the TMP/Gel displayed significantly lower-than-normal levels of pro-inflammatory and osteoclastogenic genes. Finally, the in vivo results indicated that, compared with the cryogel with no anti-inflammatory effect, the TMP/Gel significantly enhanced both the quality and quantity of newly formed bone, demonstrating the importance of combining anti-inflammation with osteoinduction. CONCLUSION: Collectively, these findings suggest our nanoparticle-hydrogel composite could be an effective tool to regulate complex events within the bone healing process.

18.
Microvasc Res ; 84(1): 1-8, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22546563

RESUMO

Stem cell transplantation can induce neovascularization. Regenerated blood vessels should remain stable for a long-term period in order to function as new blood vessels in ischemic tissues. Here we show that local delivery of FGF2 enhances the long-term (12weeks) angiogenic efficacy of human adipose-derived stem cells (hADSCs) implanted into mouse ischemic hindlimbs. Following transplantation of hADSCs into ischemic hindlimbs of mice, hADSC viability was significantly higher in the hADSC+FGF2 group at 4 and 12weeks post-transplantation than in the hADSC only group. Furthermore, hADSCs produced higher levels of angiogenic growth factors (i.e., fibroblast growth factor 2, vascular endothelial growth factor, hepatocyte growth factor, and platelet-derived growth factor) at both time points. As a result, the density of arterioles in the ischemic hindlimb muscle was significantly higher in the hADSC+FGF2 group than in either hADSC or FGF2 only group at both time points. The number of arterioles with larger diameters was significantly greater in the hADSC+FGF2 group than in the other groups at 12weeks, and increased in the hADSC+FGF2 group as the time period increased from 4weeks to 12weeks post-transplantation. This suggests that FGF2 delivery to hADSC transplantation sites enhances long-term angiogenic efficacy of hADSCs transplanted into ischemic tissues.


Assuntos
Tecido Adiposo/citologia , Fatores de Crescimento de Fibroblastos/administração & dosagem , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Neovascularização Fisiológica/fisiologia , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/patologia , Biomarcadores/metabolismo , Sobrevivência Celular , Células Cultivadas , Feminino , Fatores de Crescimento de Fibroblastos/metabolismo , Membro Posterior/patologia , Isquemia/terapia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/ultraestrutura , Camundongos , Camundongos Nus , Microscopia Eletrônica de Varredura , Músculos/irrigação sanguínea , Músculos/patologia , Transplante Heterólogo
19.
Biomacromolecules ; 13(7): 2020-8, 2012 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-22617001

RESUMO

Most polymeric vascular prosthetic materials have low patency rate for replacement of small diameter vessels (<5 mm), mainly due to failure to generate healthy endothelium. In this study, we present polydopamine-mediated immobilization of growth factors on the surface of polymeric materials as a versatile tool to modify surface characteristics of vascular grafts potentially for accelerated endothelialization. Polydopamine was deposited on the surface of biocompatible poly(L-lactide-co-ε-caprolactone) (PLCL) elastomer, on which vascular endothelial growth factor (VEGF) was subsequently immobilized by simple dipping. Surface characteristics and composition were investigated by using scanning electron microscopy, atomic force microscopy, and X-ray photoelectron spectroscopy. Immobilization of VEGF on the polydopamine-deposited PLCL films was effective (19.8 ± 0.4 and 197.4 ± 19.7 ng/cm(2) for DPv20 and DPv200 films, respectively), and biotin-mediated labeling of immobilized VEGF revealed that the fluorescence intensity increased as a function of the concentration of VEGF solution. The effect of VEGF on adhesion of HUVECs was marginal, which may have been masked by polydopamine layer that also enhanced cell adhesion. However, VEGF-immobilized substrate significantly enhanced proliferation of HUVECs for over 7 days of in vitro culture and also improved their migration. In addition, immobilized VEGF supported robust cell to cell interactions with strong expression of CD 31 marker. The same process was effective for immobilization of basic fibroblast growth factor, demonstrating the robustness of polydopamine layer for secondary ligation of growth factors as a simple and novel surface modification strategy for vascular graft materials.


Assuntos
Prótese Vascular , Proteínas Imobilizadas/química , Fator A de Crescimento do Endotélio Vascular/química , Animais , Bivalves , Adesão Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Indóis/química , Microscopia de Força Atômica , Microscopia Eletrônica de Varredura , Espectroscopia Fotoeletrônica , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Poliésteres/química , Polímeros/química , Propriedades de Superfície , Molhabilidade
20.
J Mater Chem B ; 10(14): 2649-2660, 2022 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-35024722

RESUMO

Multicellular tumor spheroids (MCTSs) are attractive for drug screening before animal tests because they emulate an in vivo microenvironment. The permeability of the MCTSs and tumor tissues towards the candidate drugs is not sufficient even though the drugs can penetrate monolayer cultured cells; therefore, nanocarriers are required to enhance permeability and deliver drugs. In this study, we prepared zwitterionic polymers of sulfobetaine methacrylates and (meth)acrylamides with or without hydroxy groups between the zwitterions to serve as highly permeable nanocarriers. In the sulfobetaine polymers, poly(2-hydroxy-3-((3-methacrylamidopropyl)dimethylammonio)propane-1-sulfonate), P(OH-MAAmSB), the hydroxy group containing methacrylamide polymer exhibited little cytotoxicity and membrane translocation ability against monolayer cultured cells. Moreover, the excellent permeability of the hepatocyte MCTS enabled P(OH-MAAmSB) to permeate it and reach the center region (∼325 µm in diameter) at approximately 150 s, although poly(trimethyl-2-methacroyloxyethylammonium), a cationic polymer, penetrated just 1 to 2 layers from the periphery. The superior permeability of P(OH-MAAmSB) might be due to its good solubility and side chain conformation. P(OH-MAAmSB) is a promising nanocarrier with membrane translocation and permeability.


Assuntos
Neoplasias , Polímeros , Animais , Betaína/análogos & derivados , Permeabilidade , Polímeros/química , Esferoides Celulares , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA