Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 220
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Clin Periodontol ; 50(2): 200-219, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36110056

RESUMO

AIM: To investigate the potential of an ultrashort aromatic peptide hydrogelator integrated with hyaluronic acid (HA) to serve as a scaffold for bone regeneration. MATERIALS AND METHODS: Fluorenylmethyloxycarbonyl-diphenylalanine (FmocFF)/HA hydrogel was prepared and characterized using microscopy and rheology. Osteogenic differentiation of MC3T3-E1 preosteoblasts was investigated using Alizarin red, alkaline phosphatase and calcium deposition assays. In vivo, 5-mm-diameter calvarial critical-sized defects were prepared in 20 Sprague-Dawley rats and filled with either FmocFF/HA hydrogel, deproteinized bovine bone mineral, FmocFF/Alginate hydrogel or left unfilled. Eight weeks after implantation, histology and micro-computed tomography analyses were performed. Immunohistochemistry was performed in six rats to assess the hydrogel's immunomodulatory effect. RESULTS: A nanofibrous FmocFF/HA hydrogel with a high storage modulus of 46 KPa was prepared. It supported osteogenic differentiation of MC3T3-E1 preosteoblasts and facilitated calcium deposition. In vivo, the hydrogel implantation resulted in approximately 93% bone restoration. It induced bone deposition not only around the margins, but also generated bony islets along the defect. Elongated M2 macrophages lining at the periosteum-hydrogel interface were observed 1 week after implantation. After 3 weeks, these macrophages were dispersed through the regenerating tissue surrounding the newly formed bone. CONCLUSIONS: FmocFF/HA hydrogel can serve as a cell-free, biomimetic, immunomodulatory scaffold for bone regeneration.


Assuntos
Ácido Hialurônico , Hidrogéis , Ratos , Animais , Bovinos , Hidrogéis/farmacologia , Hidrogéis/química , Ácido Hialurônico/farmacologia , Ácido Hialurônico/uso terapêutico , Osteogênese , Microtomografia por Raio-X , Cálcio/farmacologia , Ratos Sprague-Dawley , Regeneração Óssea , Periósteo , Alicerces Teciduais/química
2.
Int J Mol Sci ; 23(9)2022 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-35562890

RESUMO

Tissue engineering aims to repair, restore, and/or replace tissues in the human body as an alternative to grafts and prostheses. Biomaterial scaffolds can be utilized to provide a three-dimensional microenvironment to facilitate tissue regeneration. Previously, we reported that scaffold pore size influences vascularization and extracellular matrix composition both in vivo and in vitro, to ultimately influence tissue phenotype for regenerating cranial suture and bone tissues, which have markedly different tissue properties despite similar multipotent stem cell populations. To rationally design biomaterials for specific cell and tissue fate specification, it is critical to understand the molecular processes governed by cell-biomaterial interactions, which guide cell fate specification. Building on our previous work, in this report we investigated the hypothesis that scaffold pore curvature, the direct consequence of pore size, modulates the differentiation trajectory of mesenchymal stem cells (MSCs) through alterations in the cytoskeleton. First, we demonstrated that sufficiently small pores facilitate cell clustering in subcutaneous explants cultured in vivo, which we previously reported to demonstrate stem tissue phenotype both in vivo and in vitro. Based on this observation, we cultured cell-scaffold constructs in vitro to assess early time point interactions between cells and the matrix as a function of pore size. We demonstrate that principle curvature directly influences nuclear aspect and cell aggregation in vitro. Scaffold pores with a sufficiently low degree of principle curvature enables cell differentiation; pharmacologic inhibition of actin cytoskeleton polymerization in these scaffolds decreased differentiation, indicating a critical role of the cytoskeleton in transducing cues from the scaffold pore microenvironment to the cell nucleus. We fabricated a macropore model, which allows for three-dimensional confocal imaging and demonstrates that a higher principle curvature facilitates cell aggregation and the formation of a potentially protective niche within scaffold macropores which prevents MSC differentiation and retains their stemness. Sufficiently high principle curvature upregulates yes-associated protein (YAP) phosphorylation while decreased principle curvature downregulates YAP phosphorylation and increases YAP nuclear translocation with subsequent transcriptional activation towards an osteogenic differentiation fate. Finally, we demonstrate that the inhibition of the YAP/TAZ pathway causes a defect in differentiation, while YAP/TAZ activation causes premature differentiation in a curvature-dependent way when modulated by verteporfin (VP) and 1-oleyl-lysophosphatidic acid (LPA), respectively, confirming the critical role of biomaterials-mediated YAP/TAZ signaling in cell differentiation and fate specification. Our data support that the principle curvature of scaffold macropores is a critical design criterion which guides the differentiation trajectory of mesenchymal stem cells' scaffolds. Biomaterial-mediated regulation of YAP/TAZ may significantly contribute to influencing the regenerative outcomes of biomaterials-based tissue engineering strategies through their specific pore design.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Materiais Biocompatíveis/metabolismo , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Engenharia Tecidual
3.
Adv Funct Mater ; 30(21)2020 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-33071711

RESUMO

Myocardial infarction (heart attack) is the number one killer of heart patients. Existing treatments for heart attack do not address the underlying problem of cardiomyocyte (CM) loss and cannot regenerate the myocardium. Introducing exogenous cardiac cells is required for heart regeneration due to the lack of resident progenitor cells and very limited proliferative potential of adult CMs. Poor retention of transplanted cells is the critical bottleneck of heart regeneration. Here, we report the invention of a poly(l-lactic acid)-b-poly(ethylene glycol)-b-poly(N-Isopropylacrylamide) copolymer and its self-assembly into nanofibrous gelling microspheres (NF-GMS). The NF-GMS undergo thermally responsive transition to form not only a 3D hydrogel after injection in vivo, but also exhibit architectural and structural characteristics mimicking the native extracellular matrix (ECM) of nanofibrous proteins and gelling proteoglycans or polysaccharides. By integrating the ECM-mimicking features, injectable form, and the capability of maintaining 3D geometry after injection, the transplantation of hESC-derived CMs carried by NF-GMS led to a striking 10-fold graft size increase over direct CM injection in an infarcted rat model, which is the highest reported engraftment to date. Furthermore, NF-GMS carried CM transplantation dramatically reduced infarct size, enhanced integration of transplanted CMs, stimulated vascularization in the infarct zone, and led to a substantial recovery of cardiac function. The NF-GMS may also serve as advanced injectable and integrative biomaterials for cell/biomolecule delivery in a variety of biomedical applications.

4.
Orthod Craniofac Res ; 23(1): 35-43, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31593373

RESUMO

OBJECTIVE: The goal of this study was to investigate potential negative sequelae of orthodontic force application ±delivery of an osteoclast inhibitor, recombinant osteoprotegerin protein (OPG-Fc), on periodontal tissues. SETTING AND SAMPLE POPULATION: Sprague Dawley rats from a commercial supplier were investigated in a laboratory setting. MATERIALS AND METHODS: Rats were randomly divided into four groups (n = 7 each): one group with no orthodontic appliances and injected once prior to the experimental period with empty polymer microspheres, one group with orthodontic appliances and injected once with empty microspheres, one group with orthodontic appliances and injected once with polymer microspheres containing 1 mg/kg of OPG-Fc, and one group with orthodontic appliances and injected with non-encapsulated 5 mg/kg of OPG-Fc every 3 days during the experimental period. The animals were euthanized after 28 days of tooth movement for histomorphometric analyses. RESULTS: Root resorption, PDL area and widths were similar in animals without appliances and animals with appliances plus high-dose OPG-Fc. PDL blood vessels were compressed and decreased in number in all animals that received orthodontic appliances, regardless of OPG-Fc. Hyalinization was significantly increased only in animals with orthodontic appliances plus multiple injections of 5 mg/kg non-encapsulated OPG-Fc when compared to animals without appliances. CONCLUSIONS: Results of this study indicate that while pharmacological modulation of tooth movement through osteoclast inhibition is feasible when delivered in a locally controlled low-dose manner, high-dose levels that completely prevent tooth movement through bone may decrease local blood flow and increase the incidence of hyalinization.


Assuntos
Reabsorção da Raiz , Técnicas de Movimentação Dentária , Animais , Osteoclastos , Ligamento Periodontal , Ratos , Ratos Sprague-Dawley , Raiz Dentária
5.
Nano Lett ; 19(11): 7918-7926, 2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31645103

RESUMO

Intracellular protein-protein interactions (PPIs) are a vital and yet underexploited class of therapeutic targets for their crucial roles in cellular processes and involvement in disease initiation and progression. Although some successful chemistry and nanotechnologies have been introduced into peptide PPI modulators to allow cell and tissue permeability, significant challenges remain with regard to the efficient and precise modulation of PPIs within specific cells of diseased tissues, such as solid tumors. Herein, an intratumoral transformable hierarchical framework, termed iPLF, was fabricated via a two-step self-assembly between peptides and lanthanide-doped nanocrystals. In this proof-of-concept study, using NanoEL effect, TME response, and tumor marker targeting, iPLF in vivo delivered the p53-MDM2 modulator DPMI into tumor cells and ß-catenin-Bcl9 modulator Bcl9p into tumor stem cells. This crafted programmed nanomedicine with triple-stage delivery and responsiveness accurately modulated the specific intracellular protein-protein interactions, resulting in the suppression of tumor growth and metastasis in vivo, while maintaining a highly favorable safety profile. iPLF reached the goal of accurate, potent, and hazard-free intracellular PPI modulation, thereby providing a means to improve current knowledge of PPI networks and a novel therapeutic strategy for a great variety of diseases.


Assuntos
Antineoplásicos/farmacologia , Elementos da Série dos Lantanídeos/farmacologia , Neoplasias/tratamento farmacológico , Peptídeos/farmacologia , Mapas de Interação de Proteínas/efeitos dos fármacos , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Preparações de Ação Retardada/química , Desenho de Fármacos , Células HCT116 , Humanos , Elementos da Série dos Lantanídeos/química , Elementos da Série dos Lantanídeos/uso terapêutico , Camundongos , Nanomedicina , Nanopartículas/química , Neoplasias/metabolismo , Neoplasias/patologia , Peptídeos/química , Peptídeos/uso terapêutico , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , beta Catenina/metabolismo
6.
Int J Mol Sci ; 21(2)2020 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-31963842

RESUMO

Mammalian telomere lengths are primarily regulated by telomerase, a ribonucleoprotein consisting of a reverse transcriptase (TERT) and an RNA subunit (TERC). TERC is constitutively expressed in all cells, whereas TERT expression is temporally and spatially regulated, such that in most adult somatic cells, TERT is inactivated and telomerase activity is undetectable. Most tumor cells activate TERT as a mechanism for preventing progressive telomere attrition to achieve proliferative immortality. Therefore, inactivating TERT has been considered to be a promising means of cancer therapy. Here we applied the CRISPR/Cas9 gene editing system to target the TERT gene in cancer cells. We report that disruption of TERT severely compromises cancer cell survival in vitro and in vivo. Haploinsufficiency of TERT in tumor cells is sufficient to result in telomere attrition and growth retardation in vitro. In vivo, TERT haploinsufficient tumor cells failed to form xenograft after transplantation to nude mice. Our work demonstrates that gene editing-mediated TERT knockout is a potential therapeutic option for treating cancer.


Assuntos
Técnicas de Inativação de Genes/métodos , Telomerase/genética , Telomerase/metabolismo , Neoplasias do Colo do Útero/patologia , Animais , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Feminino , Haploinsuficiência , Células HeLa , Humanos , Mutação INDEL , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/metabolismo
7.
Adv Funct Mater ; 29(10)2019 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-32982625

RESUMO

Clinical translation of therapeutic peptides, particularly those targeting intracellular protein-protein interactions (PPIs), has been hampered by their inefficacious cellular internalization in diseased tissue. Therapeutic peptides engineered into nanostructures with stable spatial architectures and smart disease targeting ability may provide a viable strategy to overcome the pharmaceutical obstacles of peptides. This study describes a strategy to assemble therapeutic peptides into a stable peptide-Au nanohybrid, followed by further self-assembling into higher-order nanoclusters with responsiveness to tumor microenvironment. As a proof of concept, an anticancer peptide termed ß-catenin/Bcl9 inhibitors is copolymerized with gold ion and assembled into a cluster of nanohybrids (pCluster). Through a battery of in vitro and in vivo tests, it is demonstrated that pClusters potently inhibit tumor growth and metastasis in several animal models through the impairment of the Wnt/ß-catenin pathway, while maintaining a highly favorable biosafety profile. In addition, it is also found that pClusters synergize with the PD1/PD-L1 checkpoint blockade immunotherapy. This new strategy of peptide delivery will likely have a broad impact on the development of peptide-derived therapeutic nanomedicine and reinvigorate efforts to discover peptide drugs that target intracellular PPIs in a great variety of human diseases, including cancer.

8.
Small ; 15(12): e1900046, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30786150

RESUMO

Developing injectable nanocomposite conductive hydrogel dressings with multifunctions including adhesiveness, antibacterial, and radical scavenging ability and good mechanical property to enhance full-thickness skin wound regeneration is highly desirable in clinical application. Herein, a series of adhesive hemostatic antioxidant conductive photothermal antibacterial hydrogels based on hyaluronic acid-graft-dopamine and reduced graphene oxide (rGO) using a H2 O2 /HPR (horseradish peroxidase) system are prepared for wound dressing. These hydrogels exhibit high swelling, degradability, tunable rheological property, and similar or superior mechanical properties to human skin. The polydopamine endowed antioxidant activity, tissue adhesiveness and hemostatic ability, self-healing ability, conductivity, and NIR irradiation enhanced in vivo antibacterial behavior of the hydrogels are investigated. Moreover, drug release and zone of inhibition tests confirm sustained drug release capacity of the hydrogels. Furthermore, the hydrogel dressings significantly enhance vascularization by upregulating growth factor expression of CD31 and improve the granulation tissue thickness and collagen deposition, all of which promote wound closure and contribute to a better therapeutic effect than the commercial Tegaderm films group in a mouse full-thickness wounds model. In summary, these adhesive hemostatic antioxidative conductive hydrogels with sustained drug release property to promote complete skin regeneration are an excellent wound dressing for full-thickness skin repair.


Assuntos
Antibacterianos/farmacologia , Hemostáticos/farmacologia , Hidrogéis/química , Injeções , Fototerapia , Regeneração/efeitos dos fármacos , Pele/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Adesividade , Animais , Antioxidantes/análise , Linhagem Celular , Preparações de Ação Retardada , Dopamina/química , Liberação Controlada de Fármacos , Condutividade Elétrica , Grafite/química , Hemólise/efeitos dos fármacos , Ácido Hialurônico/química , Hipertermia Induzida , Camundongos , Oxirredução , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Reologia , Fator de Necrose Tumoral alfa/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 38(9): 2191-2197, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30026272

RESUMO

Objective- To identify the transcription factors that could contribute to direct reprogramming of fibroblasts toward smooth muscle cell fate. Approach and Results- We screened various combinations of transcription factors, including Myocd (myocardin), Mef2C (myocyte enhancer factor 2C), Mef2B (myocyte enhancer factor 2B), Mkl1 (MKL [megakaryoblastic leukemia]/Myocd-like 1), Gata4 (GATA-binding protein 4), Gata5 (GATA-binding protein 5), Gata6 (GATA-binding protein 6), Ets1 (E26 avian leukemia oncogene 1, 5' domain), and their corresponding carboxyterminal fusions to the transactivation domain of MyoD (myogenic differentiation 1)-indicated by *-for their effects on reprogramming mouse embryonic fibroblasts and human adult dermal fibroblasts to the smooth muscle cell fate as determined by the expression of specific markers. The combination of 3 transcription factors, Myocd (or Myocd*) with Mef2C (or Mef2C*) and Gata6, was the most efficient in enhancing the expression of smooth muscle marker genes and decreasing fibroblast gene expression. Additionally, the derived induced smooth muscle-like cells showed a contractile phenotype in response to carbachol. Conclusions- Combination of Myocd and Gata6 with Mef2C* (MG2*) could sufficiently and efficiently direct differentiation of mouse embryonic and human dermal fibroblasts into induced smooth muscle-like cells, thus opening new opportunities for disease modeling, tissue engineering, and personalized medicine.


Assuntos
Técnicas de Reprogramação Celular/métodos , Fibroblastos/citologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Fatores de Transcrição , Animais , Carbacol/farmacologia , Diferenciação Celular , Células Cultivadas , Reprogramação Celular , Embrião de Mamíferos , Expressão Gênica , Humanos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/efeitos dos fármacos , Fenótipo , Pele/citologia
10.
Stem Cells ; 35(2): 351-361, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27571517

RESUMO

Vascular smooth muscle cells (VSMCs) derived from cardiovascular progenitor cell (CVPC) lineage populate the tunica media of the aortic root. Understanding differentiation of VSMCs from CVPC will further our understanding of the molecular mechanisms contributing to aortic root aneurysms, and thus, facilitate the development of novel therapeutic agents to prevent this devastating complication. It is established that the yes-associated protein (YAP) and Hippo pathway is important for VSMC proliferation and phenotype switch. To determine the role of YAP in differentiation of VSMCs from CVPCs, we utilized the in vitro monolayer lineage specific differentiation method by differentiating human embryonic stem cells into CVPCs, and then, into VSMCs. We found that expression of YAP decreased during differentiation of VSMC from CVPCs. Overexpression of YAP attenuated expression of VSMC contractile markers and impaired VSMC function. Knockdown of YAP increased expression of contractile proteins during CVPC-VSMCs differentiation. Importantly, expression of YAP decreased transcription of myocardin during this process. Overexpression of YAP in PAC1 SMC cell line inhibited luciferase activity of myocardin proximal promoter in a dose dependent and NKX2.5 dependent manners. YAP protein interacted with NKX2.5 protein and inhibited binding of NKX2.5 to the 5'-proximal promoter region of myocardin in CVPC-derived VSMCs. In conclusion, YAP negatively regulates differentiation of VSMCs from CVPCs by decreasing transcription of myocardin in a NKX2.5-dependent manner. Stem Cells 2017;35:351-361.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Diferenciação Celular/genética , Linhagem da Célula , Miócitos Cardíacos/citologia , Miócitos de Músculo Liso/citologia , Proteínas Nucleares/genética , Fosfoproteínas/metabolismo , Células-Tronco/citologia , Transativadores/genética , Transcrição Gênica , Biomarcadores/metabolismo , Linhagem Celular , Técnicas de Silenciamento de Genes , Proteína Homeobox Nkx-2.5/metabolismo , Humanos , Modelos Biológicos , Músculo Liso Vascular/citologia , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Transativadores/metabolismo , Fatores de Transcrição , Proteínas de Sinalização YAP
11.
Biomacromolecules ; 19(6): 1764-1782, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29684268

RESUMO

Electrically conducting polymers such as polyaniline, polypyrrole, polythiophene, and their derivatives (mainly aniline oligomer and poly(3,4-ethylenedioxythiophene)) with good biocompatibility find wide applications in biomedical fields including bioactuators, biosensors, neural implants, drug delivery systems, and tissue engineering scaffolds. This review focuses on these conductive polymers for tissue engineering applications. Conductive polymers exhibit promising conductivity as bioactive scaffolds for tissue regeneration, and their conductive nature allows cells or tissue cultured on them to be stimulated by electrical signals. However, their mechanical brittleness and poor processability restrict their application. Therefore, conductive polymeric composites based on conductive polymers and biocompatible biodegradable polymers (natural or synthetic) were developed. The major objective of this review is to summarize the conductive biomaterials used in tissue engineering including conductive composite films, conductive nanofibers, conductive hydrogels, and conductive composite scaffolds fabricated by various methods such as electrospinning, coating, or deposition by in situ polymerization. Furthermore, recent progress in tissue engineering applications using these conductive biomaterials including bone tissue engineering, muscle tissue engineering, nerve tissue engineering, cardiac tissue engineering, and wound healing application are discussed in detail.


Assuntos
Materiais Biocompatíveis/química , Polímeros/química , Engenharia Tecidual/métodos , Animais , Materiais Biocompatíveis/farmacologia , Humanos , Hidrogéis/química , Nanofibras/química , Polímeros/síntese química , Engenharia Tecidual/instrumentação , Alicerces Teciduais/química , Cicatrização
12.
Biomacromolecules ; 18(9): 2808-2819, 2017 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-28792734

RESUMO

The key factor in skeletal muscle tissue engineering is regeneration of the functional skeletal muscles. Materials that could promote the myoblast proliferation and myogenic differentiation are promising candidates in skeletal muscle tissue engineering. Herein, we developed an elastic conductive poly(ethylene glycol)-co-poly(glycerol sebacate) (PEGS) grafted aniline pentamer (AP) copolymer that could promote the formation of myotubes by differentiating the C2C12 myoblast cells. The results of hydration behavior and water contact angle suggested that by adjusting the poly(ethylene glycol) (PEG) and AP content, this film showed a proper surface hydrophilicity for cell attachment. Additionally, these films showed tunable conductivity and mechanical properties that can be altered by changing the AP content. The maximum conductivity of the films was 1.84 × 10-4 S/cm and the Young's modulus of these films ranged from 14.58 ± 1.35 MPa to 24.62 ± 0.61 MPa. Our findings indicate that the PEGS-AP films promote the proliferation and myogenic differentiation of C2C12 cells, suggesting that they are promising biomaterials for skeletal muscle tissue engineering.


Assuntos
Mioblastos Esqueléticos/citologia , Regeneração , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Compostos de Anilina/química , Animais , Linhagem Celular , Módulo de Elasticidade , Condutividade Elétrica , Camundongos , Desenvolvimento Muscular , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/fisiologia , Poliésteres/química , Alicerces Teciduais/efeitos adversos
13.
J Cell Biochem ; 117(7): 1556-67, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26579618

RESUMO

Anabolic actions of PTH in bone involve increased deposition of mineralizing matrix. Regulatory feedback of the process may be important to maintain calcium homeostasis and, in turn, calcium may inform the process. This investigation clarified the role of calcium availability and the calcium sensing receptor (CaSR) in the anabolic actions of PTH. CaSR function promoted osteoblastic cell numbers, with lower cell numbers in post-confluent cultures of primary calvarial cells from Col1-CaSR knock-out (KO) mice, and for calvarial cells from wild-type (WT) mice treated with a calcilytic. Increased apoptosis of calvarial cells with calcilytic treatment suggested CaSR is critical for protection against stage-dependent cell death. Whole and cortical, but not trabecular, bone parameters were significantly lower in Col1-CaSR KO mice versus WT littermates. Intact Col1-CaSR KO mice had lower serum P1NP levels relative to WT. PTH treatment displayed anabolic actions in WT and, to a lesser degree, KO mice, and rescued the lower P1NP levels in KO mice. Furthermore, PTH effects on whole tibiae were inhibited by osteoblast-specific CaSR ablation. Vertebral body implants (vossicles) from untreated Col1-CaSR KO and WT mice had similar bone volumes after 4 weeks of implantation in athymic mice. These findings suggest that trabecular bone formation can occur independently of the CaSR, and that the CaSR plays a collaborative role in the PTH anabolic effects on bone. J. Cell. Biochem. 117: 1556-1567, 2016. © 2015 Wiley Periodicals, Inc.


Assuntos
Osteogênese/fisiologia , Hormônio Paratireóideo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Crânio/metabolismo , Animais , Cálcio , Sobrevivência Celular , Camundongos , Camundongos Knockout , Camundongos Nus , Osteoclastos , Hormônio Paratireóideo/genética , Fragmentos de Peptídeos/metabolismo , Pró-Colágeno/metabolismo , Receptores de Detecção de Cálcio , Receptores Acoplados a Proteínas G/genética
14.
Adv Funct Mater ; 25(3): 350-360, 2015 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-26069467

RESUMO

Injectable microspheres are attractive stem cell carriers for minimally invasive procedures. For tissue regeneration, the microspheres need to present the critical cues to properly direct stem cell differentiation. In natural extracellular matrix (ECM), growth factors (GFs) and collagen nanofibers provide critical chemical and physical cues. However, there have been no reported technologies that integrate synthetic nanofibers and GFs into injectable microspheres. In this study, we synthesized functional nanofibrous hollow microspheres (FNF-HMS), which can covalently bind GF-mimicking peptides. Two different GF-mimicking peptides, Transforming Growth Factor-ß1 mimicking peptide Cytomodulin (CM) and Bone Morphogenetic Protein-2 mimicking peptide P24, were separately conjugated onto the FNF-HMS to induce distinct differentiation pathways of rabbit bone marrow-derived mesenchymal stem cells (BMSCs). While no existing biomaterials were reported to successfully deliver CM to induce chondrogenesis, the developed FNF-HMS were shown to effectively present CM to BMSCs and successfully induced their chondrogenesis for cartilage formation in both in vitro and in vivo studies. In addition, P24 was conjugated onto the newly developed FNF-HMS and was capable of retaining its bioactivity and inducing ectopic bone formation in nude mice. These results demonstrate that the novel FNF-HMS can effectively deliver GF-mimicking peptides to modulate stem cell fate and tissue regeneration.

15.
Macromol Rapid Commun ; 36(19): 1735-41, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26247175

RESUMO

Nano- and microsized structures are of central importance to advanced materials and nanotechnologies, which have tremendously impacted both biomedical and physical sciences. Herein, novel emulsification and thermally induced phase separation (TIPS) techniques to fabricate linear polymers into nanofibrous hollow objects are reported for the first time. Through manipulating the emulsification conditions, the evolution of the emulsion structure can be controlled and nanofibrous hollow microspheres with a controllable opening size and nano-fibrous shells can be fabricated. Through adjusting the rheological properties of the emulsions, nanofibrous hollow discs are also created. A new mechanistic hypotheses of the nanofibrous hollow object formation is proposed: the nano- and microscaled structures are independently determined by TIPS and the emulsification process, respectively. Guided by this theory, the nanofiber formation conditions for two further additional polymers (polyacrylonitrile and Nylon) under TIPS are identified, and solid/nanofibrous non-hollow/hollow microspheres are created from these two additional polymers under TIPS and emulsification for the first time. Therefore, the developed strategy is applicable to various polymer systems, and can broadly impact nano- and microfabrication technologies.


Assuntos
Microesferas , Nanofibras/química , Emulsões/química , Nanofibras/ultraestrutura , Polímeros/química , Tensoativos/química , Temperatura
16.
Biomacromolecules ; 15(9): 3246-52, 2014 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-25102223

RESUMO

Multifunctional injectable thermo-/pH-responsive hydrogels as release systems for the oral delivery of small molecule drugs and the local delivery of protein are presented. The injectable interpenetrating polymer network (IPN) hydrogels based on poly(ethylene glycol) methacrylate, N-isopropylacrylamide, and methacrylated alginate were prepared by using ammonium persulfate (APS) and N,N,N',N'-tetramethylethylenediamine (TEMED) as a redox initiator system at body temperature, and the obtained hydrogels overcame the instability of calcium cross-linked alginate hydrogels under physiological conditions. The hydrogels showed good mechanical strength by rheometer and exhibited temperature and pH sensitivity by a swelling test. Diclofenac sodium (DCS) as a model for small molecule water-soluble anti-inflammatory drugs and bovine serum albumin (BSA) as a model for protein drugs were encapsulated in situ in the hydrogel. The DCS and BSA release results indicated that these hydrogels, as carriers, have great potential for use in the oral delivery of small molecule drugs and for long-term localized protein release. Furthermore, the cytotoxicity of these hydrogels was studied via live/dead viability and alamarBlue assays using adipose tissue-derived mesenchymal stem cells.


Assuntos
Alginatos , Anti-Inflamatórios não Esteroides , Diclofenaco , Portadores de Fármacos , Hidrogéis , Células-Tronco Mesenquimais/metabolismo , Soroalbumina Bovina , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Alginatos/síntese química , Alginatos/química , Alginatos/farmacocinética , Alginatos/farmacologia , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacocinética , Anti-Inflamatórios não Esteroides/farmacologia , Bovinos , Preparações de Ação Retardada/síntese química , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacocinética , Preparações de Ação Retardada/farmacologia , Diclofenaco/química , Diclofenaco/farmacocinética , Diclofenaco/farmacologia , Portadores de Fármacos/síntese química , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacologia , Ácido Glucurônico/síntese química , Ácido Glucurônico/química , Ácido Glucurônico/farmacocinética , Ácido Glucurônico/farmacologia , Ácidos Hexurônicos/síntese química , Ácidos Hexurônicos/química , Ácidos Hexurônicos/farmacocinética , Ácidos Hexurônicos/farmacologia , Hidrogéis/síntese química , Hidrogéis/química , Hidrogéis/farmacocinética , Hidrogéis/farmacologia , Concentração de Íons de Hidrogênio , Células-Tronco Mesenquimais/citologia , Metacrilatos/química , Coelhos , Soroalbumina Bovina/química , Soroalbumina Bovina/farmacocinética , Soroalbumina Bovina/farmacologia
17.
Macromol Biosci ; 24(6): e2300540, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38456554

RESUMO

The scaffold is a porous three-dimensional (3D) material that supports cell growth and tissue regeneration. Such 3D structures should be generated with simple techniques and nontoxic ingredients to mimic bio-environment and facilitate tissue regeneration. In this work, simple but powerful techniques are demonstrated for the fabrication of lamellar and honeycomb-mimic scaffolds with poly(L-lactic acid). The honeycomb-mimic scaffolds with tunable pore size ranging from 70 to 160 µm are fabricated by crystal needle-guided thermally induced phase separation in a directional freezing apparatus. The compressive modulus of the honeycomb-mimic scaffold is ≈4 times higher than that of scaffold with randomly oriented pore structure. The fabricated honeycomb-mimic scaffold exhibits a hierarchical structure from nanofibers to micro-/macro-tubular structures. Pre-osteoblast MC3T3-E1 cells cultured on the honeycomb-mimic nanofibrous scaffolds exhibit an enhanced osteoblastic phenotype, with elevated expression levels of osteogenic marker genes, than those on either porous lamellar scaffolds or porous scaffolds with randomly oriented pores. The advanced techniques for the fabrication of the honeycomb-mimic structure may potentially be used for a wide variety of advanced functional materials.


Assuntos
Nanofibras , Osteoblastos , Poliésteres , Alicerces Teciduais , Alicerces Teciduais/química , Nanofibras/química , Camundongos , Animais , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoblastos/efeitos dos fármacos , Poliésteres/química , Porosidade , Linhagem Celular , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Engenharia Tecidual/métodos , Osteogênese/efeitos dos fármacos
18.
J Biomed Mater Res A ; 112(6): 852-865, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38192179

RESUMO

Stimuli-responsive domains capable of releasing loaded molecules, "on-demand," have garnered increasing attention due to their enhanced delivery, precision targeting, and decreased adverse effects. The development of an on-demand delivery system that can be easily triggered by dental clinicians might have major roles in dental and oral tissue engineering. A series of random graft poly(NIPAm-co-HEMA-Lactate) copolymers were synthesized using 95:5, 85:5, 60:40, and 40:60 ratios of thermosensitive NIPAm and HEMA-poly lactate respectively then electrospun to produce nanofibrous scaffolds loaded with bovine serum albumin (BSA). Cumulative BSA release was assessed at 25C and 37°C. To appraise the use of scaffolds as on-demand delivery systems, they were subjected to thermal changes in the form cooling and warming cycles during which BSA release was monitored. To confirm the triggered releasing ability of the synthesized scaffolds, the copolymer made with 60% NIPAm was selected, based on the results of the release tests, and loaded with bone morphogenetic protein-2 (BMP-2). The loaded scaffolds were placed with mesenchymal-like stem cells (iMSCs) derived from induced pluripotent stem cells (iPSCs), and subjected to temperature alterations. Then, the osteogenic differentiation of iMSCs, which might have resulted from the released protein, was evaluated after 10 days by analyzing runt-related transcription factor 2 (RUNX-2) osteogenic gene expression by the cells using real-time quantitative polymerase chain reaction (qRT-PCR). BSA release profiles showed a burst release at the beginning followed by a more linear pattern at 25°C, and a much slower release at 37°C. The release also decreased when the PNIPAm content decreased in the scaffolds. Thermal triggering led to a step-like release pattern in which the highest release was reported 30 min through the warming cycles. The iMSCs cultivated with scaffolds loaded with BMP-2 and exposed to temperature alteration showed significantly higher RUNX-2 gene expression than cells in the other experimental groups. The synthesized scaffolds are thermo-responsive and could be triggered to deliver biological biomolecules to be used in oral and dental tissue engineering. Thermal stimuli could be simulated by dental clinicians using simple means of cold therapy, for example, cold packs in intraoral accessible sites for specified times.


Assuntos
Resinas Acrílicas , Nanofibras , Osteogênese , Polímeros/farmacologia , Engenharia Tecidual/métodos , Soroalbumina Bovina/farmacologia , Ácido Láctico/farmacologia , Alicerces Teciduais
19.
J Biol Chem ; 287(18): 14598-605, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22411986

RESUMO

The Hippo-Yap (Yes-associated protein) signaling pathway has emerged as one of the critical pathways regulating cell proliferation, differentiation, and apoptosis in response to environmental and developmental cues. However, Yap1 roles in vascular smooth muscle cell (VSMC) biology have not been investigated. VSMCs undergo phenotypic switch, a process characterized by decreased gene expression of VSMC contractile markers and increased proliferation, migration, and matrix synthesis. The goals of the present studies were to investigate the relationship between Yap1 and VSMC phenotypic switch and to determine the molecular mechanisms by which Yap1 affects this essential process in VSMC biology. Results demonstrated that the expression of Yap1 was rapidly up-regulated by stimulation with PDGF-BB (a known inducer of phenotypic switch in VSMCs) and in the injured vessel wall. Knockdown of Yap1 impaired VSMC proliferation in vitro and enhanced the expression of VSMC contractile genes as well by increasing serum response factor binding to CArG-containing regions of VSMC-specific contractile genes within intact chromatin. Conversely, the interaction between serum response factor and its co-activator myocardin was reduced by overexpression of Yap1 in a dose-dependent manner. Taken together, these results indicate that down-regulation of Yap1 promotes VSMC contractile phenotype by both up-regulating myocardin expression and promoting the association of the serum response factor-myocardin complex with VSMC contractile gene promoters and suggest that the Yap1 signaling pathway is a central regulator of phenotypic switch of VSMCs.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Regulação da Expressão Gênica/fisiologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/biossíntese , Transativadores/biossíntese , Animais , Proteínas Reguladoras de Apoptose/genética , Becaplermina , Movimento Celular/fisiologia , Proliferação de Células , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas c-sis/genética , Proteínas Proto-Oncogênicas c-sis/metabolismo , Ratos , Transativadores/genética , Proteínas de Sinalização YAP
20.
Biophys Rev (Melville) ; 4(1): 011313, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37008888

RESUMO

Gene therapy and gene delivery have drawn extensive attention in recent years especially when the COVID-19 mRNA vaccines were developed to prevent severe symptoms caused by the corona virus. Delivering genes, such as DNA and RNA into cells, is the crucial step for successful gene therapy and remains a bottleneck. To address this issue, vehicles (vectors) that can load and deliver genes into cells are developed, including viral and non-viral vectors. Although viral gene vectors have considerable transfection efficiency and lipid-based gene vectors become popular since the application of COVID-19 vaccines, their potential issues including immunologic and biological safety concerns limited their applications. Alternatively, polymeric gene vectors are safer, cheaper, and more versatile compared to viral and lipid-based vectors. In recent years, various polymeric gene vectors with well-designed molecules were developed, achieving either high transfection efficiency or showing advantages in certain applications. In this review, we summarize the recent progress in polymeric gene vectors including the transfection mechanisms, molecular designs, and biomedical applications. Commercially available polymeric gene vectors/reagents are also introduced. Researchers in this field have never stopped seeking safe and efficient polymeric gene vectors via rational molecular designs and biomedical evaluations. The achievements in recent years have significantly accelerated the progress of polymeric gene vectors toward clinical applications.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA