Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Tissue Eng Part A ; 25(17-18): 1191-1201, 2019 09.
Article in English | MEDLINE | ID: mdl-31237484

ABSTRACT

IMPACT STATEMENT: A critical attribute for the long-term success of cartilage defect repair is the strong integration between the repair tissue and the surrounding native tissue. Current approaches utilized by physicians fail to achieve this attribute, leading to eventual relapse of the defect. This article demonstrates the concept of a simple, clinically viable approach for enhancing tissue integration via the combination of a safe, transient enzymatic treatment with a locally delivered, retained growth factor through an in vitro hydrogel/cartilage explant model.


Subject(s)
Cartilage/drug effects , Insulin-Like Growth Factor I/therapeutic use , Trypsin/therapeutic use , Animals , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cattle , Cell Movement/drug effects , Cell Proliferation/drug effects , Chondrocytes/cytology , Chondrocytes/drug effects , Glycosaminoglycans/metabolism , Humans , Microscopy, Confocal , Tissue Engineering
2.
ACS Synth Biol ; 8(5): 1010-1025, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30920800

ABSTRACT

Quantification of biology's central dogma (transcription and translation) is pursued by a variety of methods. Direct, immediate, and ongoing quantification of these events is difficult to achieve. Common practice is to use fluorescent or luminescent proteins to report indirectly on prior cellular events, such as turning on a gene in a genetic circuit. We present an alternative approach, PURExpress-ReAsH-Spinach In-vitro Analysis (PERSIA). PERSIA provides information on the production of RNA and protein during cell-free reactions by employing short RNA and peptide tags. Upon synthesis, these tags yield quantifiable fluorescent signal without interfering with other biochemical events. We demonstrate the applicability of PERSIA in measuring cell-free transcription, translation, and other enzymatic activity in a variety of applications: from sequence-structure-function studies, to genetic code engineering, to testing antiviral drug resistance.


Subject(s)
Cell-Free System , Protein Biosynthesis , Transcription, Genetic , Genetic Engineering/methods , HIV/enzymology , HIV Protease/genetics , HIV Protease/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , RNA, Messenger/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Spectrometry, Fluorescence , Spinacia oleracea/genetics , Ubiquitin/genetics , Ubiquitin/metabolism
4.
Tissue Eng Part A ; 22(13-14): 917-27, 2016 07.
Article in English | MEDLINE | ID: mdl-27268956

ABSTRACT

Tissue engineering approaches using growth factor-functionalized acellular scaffolds to support and guide repair driven by endogenous cells are thought to require a careful balance between cell recruitment and growth factor release kinetics. The objective of this study was to identify a growth factor combination that accelerates progenitor cell migration into self-assembling peptide hydrogels in the context of cartilage defect repair. A novel 3D gel-to-gel migration assay enabled quantification of the chemotactic impact of platelet-derived growth factor-BB (PDGF-BB), heparin-binding insulin-like growth factor-1 (HB-IGF-1), and transforming growth factor-ß1 (TGF-ß1) on progenitor cells derived from subchondral bovine trabecular bone (bone-marrow progenitor cells, BM-PCs) encapsulated in the peptide hydrogel [KLDL]3. Only the combination of PDGF-BB and TGF-ß1 stimulated significant migration of BM-PCs over a 4-day period, measured by confocal microscopy. Both PDGF-BB and TGF-ß1 were slowly released from the gel, as measured using their (125)I-labeled forms, and they remained significantly present in the gel at 4 days. In the context of augmenting microfracture surgery for cartilage repair, our strategy of delivering chemotactic and proanabolic growth factors in KLD may provide the necessary local stimulus to help increase defect cellularity, providing more cells to generate repair tissue.


Subject(s)
Bone Marrow Cells/metabolism , Cell Movement/drug effects , Insulin-Like Growth Factor I/pharmacology , Proto-Oncogene Proteins c-sis/pharmacology , Stem Cells/metabolism , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/pharmacology , Animals , Becaplermin , Bone Marrow Cells/cytology , Cattle , Stem Cells/cytology
5.
J Orthop Res ; 34(12): 2096-2105, 2016 12.
Article in English | MEDLINE | ID: mdl-26990682

ABSTRACT

A soluble form of BMP receptor type 1A (mBMPR1A-mFC) acts as an antagonist to endogenous BMPR1A and has been shown to increase bone mass in mice. The goal of this study was to examine the effects of mBMPR1A-mFC on secondary fracture healing. Treatment consisted of 10 mg/kg intraperitoneal injections of mBMPR1A-mFC twice weekly in male C57BL/6 mice. Treatment beginning at 1, 14, and 21 days post-fracture assessed receptor function during endochondral bone formation, at the onset of secondary bone formation, and during coupled remodeling, respectively. Control animals received saline injections. mBMPR1A-mFC treatment initiated on day 1 delayed cartilage maturation in the callus and resulted in large regions of fibrous tissue. Treatment initiated on day 1 also increased the amount of mineralized tissue and up-regulated many bone-associated genes (p = 0.002) but retarded periosteal bony bridging and impaired strength and toughness at day 35 (p < 0.035). Delaying the onset of treatment to day 14 or 21 partially mitigated these effects and produced evidence of accelerated coupled remodeling. These results indicate that inhibition of the BMPR1A-mediated signaling has negative effects on secondary fracture healing that are differentially manifested at different stages of healing and within different cell populations. These effects are most pronounced during the endochondral period and appear to be mediated by selective inhibition of BMPRIA signaling within the periosteum. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:2096-2105, 2016.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/administration & dosage , Bone Morphogenetic Protein Receptors, Type I/antagonists & inhibitors , Fracture Healing/drug effects , Fractures, Bone/drug therapy , Animals , Drug Evaluation, Preclinical , Male , Mice, Inbred C57BL
6.
Tissue Eng Part A ; 21(3-4): 637-46, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25231349

ABSTRACT

Heparin-binding insulin-like growth factor 1 (HB-IGF-1) is a fusion protein of IGF-1 with the HB domain of heparin-binding epidermal growth factor-like growth factor. A single dose of HB-IGF-1 has been shown to bind specifically to cartilage and to promote sustained upregulation of proteoglycan synthesis in cartilage explants. Achieving strong integration between native cartilage and tissue-engineered cartilage remains challenging. We hypothesize that if a growth factor delivered by the tissue engineering scaffold could stimulate enhanced matrix synthesis by both the cells within the scaffold and the adjacent native cartilage, integration could be enhanced. In this work, we investigated methods for adsorbing HB-IGF-1 to self-assembling peptide hydrogels to deliver the growth factor to encapsulated chondrocytes and cartilage explants cultured with growth factor-loaded hydrogels. We tested multiple methods for adsorbing HB-IGF-1 in self-assembling peptide hydrogels, including adsorption prior to peptide assembly, following peptide assembly, and with/without heparan sulfate (HS, a potential linker between peptide molecules and HB-IGF-1). We found that HB-IGF-1 and HS were retained in the peptide for all tested conditions. A subset of these conditions was then studied for their ability to stimulate increased matrix production by gel-encapsulated chondrocytes and by chondrocytes within adjacent native cartilage. Adsorbing HB-IGF-1 or IGF-1 prior to peptide assembly was found to stimulate increased sulfated glycosaminoglycan per DNA and hydroxyproline content of chondrocyte-seeded hydrogels compared with basal controls at day 10. Cartilage explants cultured adjacent to functionalized hydrogels had increased proteoglycan synthesis at day 10 when HB-IGF-1 was adsorbed, but not IGF-1. We conclude that delivery of HB-IGF-1 to focal defects in cartilage using self-assembling peptide hydrogels is a promising technique that could aid cartilage repair via enhanced matrix production and integration with native tissue.


Subject(s)
Chondrocytes/cytology , Chondrocytes/physiology , Delayed-Action Preparations/chemistry , Heparin-binding EGF-like Growth Factor/administration & dosage , Insulin-Like Growth Factor I/administration & dosage , Peptides/chemistry , Adsorption , Animals , Cattle , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Chondrocytes/drug effects , Crystallization/methods , Delayed-Action Preparations/administration & dosage , Diffusion , Drug Design , Heparin-binding EGF-like Growth Factor/chemistry , Heparin-binding EGF-like Growth Factor/genetics , Hydrogels/chemical synthesis , Insulin-Like Growth Factor I/chemistry , Insulin-Like Growth Factor I/genetics , Materials Testing , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...