Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Pediatr Hematol Oncol ; 44(5): e914-e917, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35398865

ABSTRACT

Heterozygous loss-of-function variants in the suppressor of fused protein gene (SUFU) can result in Gorlin syndrome, which is characterized by an increased frequency of basal cell carcinoma, medulloblastoma, odontogenic keratocysts, as well as other tumors. We describe a case of a 5-month-old female who presented with multiple intra-abdominal leiomyomata and was found to have a likely pathogenic splice site variant in the SUFU gene. This is the first reported case of leiomyomatosis secondary to a pathogenic SUFU variant in an infant and may represent an early, atypical presentation of Gorlin syndrome.


Subject(s)
Basal Cell Nevus Syndrome , Cerebellar Neoplasms , Leiomyomatosis , Medulloblastoma , Cerebellar Neoplasms/pathology , Female , Humans , Infant , Leiomyomatosis/complications , Leiomyomatosis/genetics , Medulloblastoma/pathology , Repressor Proteins/genetics
3.
J Adolesc Young Adult Oncol ; 10(5): 614-617, 2021 10.
Article in English | MEDLINE | ID: mdl-32960134

ABSTRACT

Biologic therapies have revolutionized the treatment of immune-mediated diseases. They are generally well tolerated; however, there are reports of malignancies associated with the use of these drugs. This case is of an adolescent with refractory Crohn's disease treated with ustekinumab, who subsequently developed Ewing's sarcoma. Patients treated with ustekinumab have reportedly developed B cell lymphoma, epithelioid sarcoma, as well as cancer of the lung, esophagus, ovary, testis, kidney, and thyroid. However, this is the first documented case of a patient treated with ustekinumab to develop Ewing sarcoma.


Subject(s)
Crohn Disease , Sarcoma, Ewing , Sarcoma , Adolescent , Crohn Disease/drug therapy , Female , Humans , Male , Sarcoma, Ewing/drug therapy , Ustekinumab/therapeutic use
4.
Orthopedics ; 40(5): e862-e867, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28817159

ABSTRACT

Little data exist to guide the treatment of unicameral bone cysts in the proximal femur. Methods of treatment include corticosteroid injections, curettage and bone grafting, and internal fixation. The authors completed a multi-institutional, retrospective review to evaluate their experience with proximal femoral unicameral bone cysts. They posed the following questions: (1) Does internal fixation reduce the risk of further procedures for the treatment of a unicameral bone cyst? (2) Is radiographic healing faster with internal fixation? Following institutional review board approval, the authors conducted a retrospective review of 36 patients treated for a unicameral bone cyst of the proximal femur at their institutions between 1974 and 2014. Medical records and radiographs were reviewed to identify patient demographics and treatment outcomes. Tumor locations included femoral neck (n=13), intertrochanteric (n=16), and subtrochanteric (n=7). Initial treatment included steroid injection (n=2), curettage and bone grafting (n=9), and internal fixation with curettage and bone grafting (n=25). Mean time was 9 months to radiographic healing and 15 months to return to full activity. The number of patients requiring additional surgeries was increased among those who did not undergo internal fixation. There was no difference in time to radiographic healing. However, time to return to normal activities was reduced if patients had received internal fixation. A significant reduction in additional procedures was observed when patients had been treated with internal fixation. Although this did not influence time to radiographic healing, patients did return to normal activities sooner. Internal fixation should be considered in the treatment of proximal femoral unicameral bone cysts. [Orthopedics. 2017; 40(5):e862-e867.].


Subject(s)
Bone Cysts/surgery , Femur/surgery , Orthopedic Procedures/methods , Adrenal Cortex Hormones , Adult , Bone Transplantation , Curettage , Female , Femur Neck/surgery , Fracture Fixation, Internal , Humans , Male , Radiography , Retrospective Studies , Treatment Outcome
5.
Case Rep Emerg Med ; 2017: 3967587, 2017.
Article in English | MEDLINE | ID: mdl-28255470

ABSTRACT

Morel-Lavallée lesions are uncommon injuries that can be associated with significant comorbidities if not detected early. Rapid diagnosis in the Emergency Department could significantly improve patient outcomes. We describe the diagnosis of such a lesion through the use of ultrasound imaging in the Emergency Department to utilize a fast, cost-effective imaging technique that does not subject the patient to radiation exposure. Our patient received surgical consultation but improved with conservative management. Ultrasound findings associated with this lesion do not require specialized equipment and should be considered when evaluating soft tissue lesions using point of care ultrasound.

6.
J Pediatr Orthop ; 37(8): e536-e542, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27137901

ABSTRACT

BACKGROUND: Adolescent idiopathic scoliosis (AIS) patients undergoing posterior spinal fusion (PSF) experience variations in their hospital care, which may lead to differences in objective and patient-reported outcomes. The purpose of this study was to demonstrate that using plan of care-educating families preoperatively and standardizing some aspects of care-would decrease time to mobility and time to discharge while maintaining pain control and patient satisfaction. METHODS: Chart review was conducted in 3 groups-preprotocol (December 2008 to December 2009, n=51), first protocol (December 2, 2009 to July 24, 2013, n=100), and second protocol (July 25, 2013 to June 1, 2014, n=39)-to track pain scores (0 to 10), time to regular diet, Foley catheter removal, epidural catheter removal, mobility, and discharge. Patient satisfaction surveys (0 to 10) were administered before discharge. Statistical analysis was performed using a 1-way analysis of variance test with Tukey post hoc analysis. RESULTS: Average pain scores were similar in all groups. Time to sitting was significantly reduced in both first protocol (27.2±9.8 h, P=1×10) and second protocol (28.4±13.6 h, P=3×10) compared with preprotocol (40.2±15.4 h). Time to discharge was significantly lower in second protocol (84.3±27.2 h, P=0.036) compared with first protocol (98.4±27.8 h). Patient satisfaction with care was significantly higher in first protocol (9.1/10, P=2×10) and second protocol (8.6/10, P=5×10) compared with preprotocol (6.5/10). CONCLUSIONS: By educating families preoperatively and standardizing portions of postoperative care in PSF for AIS, pain scores were significantly reduced while overall satisfaction remained high. Specifically, by removing the epidural and Foley catheters on postoperative day 2, time to discharge was dramatically decreased by 15 hours. The application of a multidisciplinary, evidence-driven plan of care for AIS patients undergoing PSF improves throughput and has beneficial effects on objective and patient-reported outcomes. LEVEL OF EVIDENCE: Level III-retrospective case series.


Subject(s)
Length of Stay , Patient Satisfaction , Scoliosis/psychology , Spinal Fusion/psychology , Adolescent , Female , Humans , Male , Pain Management , Patient Reported Outcome Measures , Postoperative Period , Retrospective Studies , Scoliosis/surgery , Spinal Fusion/methods , Treatment Outcome
7.
JBJS Case Connect ; 6(4): e97, 2016.
Article in English | MEDLINE | ID: mdl-29252751

ABSTRACT

CASE: A 15-year-old girl with adolescent idiopathic scoliosis with a 50° curve underwent posterior spinal fusion from T3 to T11. After discharge from the hospital, the patient reported dysphonia and dysphagia. Flexible nasendoscopy confirmed left vocal cord paresis. Stretch injury to the recurrent laryngeal nerve from the left T5 pedicle screw or intubation may have caused the vocal cord paresis. The pedicle screw was removed during revision surgery. Postsurgically, the patient demonstrated immediate and ultimately full recovery and no longer had any symptoms. CONCLUSION: To our knowledge, this is the first case report of vocal cord paresis most likely caused by pedicle screw position after posterior spinal fusion.


Subject(s)
Postoperative Complications/etiology , Scoliosis/surgery , Spinal Fusion/adverse effects , Vocal Cord Paralysis/etiology , Adolescent , Child , Female , Humans
8.
Tissue Eng Part A ; 21(3-4): 530-40, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25228401

ABSTRACT

Minimally invasive, injectable bone tissue engineering therapies offer the potential to facilitate orthopedic repair procedures, including in indications where enhanced bone regeneration is needed for complete healing. In this study, we developed a dual-phase tissue construct consisting of osteogenic (Osteo) and vasculogenic (Vasculo) components. A modular tissue engineering approach was used to create collagen/fibrin/hydroxyapatite (COL/FIB/HA) hydrogel microbeads containing embedded human bone marrow-derived mesenchymal stem cells (bmMSC). These microbeads were predifferentiated toward the osteogenic lineage in vitro for 14 days, and they were then embedded within a COL/FIB vasculogenic phase containing a coculture of undifferentiated bmMSC and human umbilical vein endothelial cells (HUVEC). In vitro studies demonstrated homogenous dispersion of microbeads within the outer phase, with endothelial network formation around the microbeads over 14 days in the coculture conditions. Subcutaneous injection into immunodeficient mice was used to investigate the ability of dual-phase (Osteo+Vasculo) and control (Osteo, Vasculo, Blank) constructs to form neovasculature and ectopic bone. Laser Doppler imaging demonstrated blood perfusion through all constructs at 1, 4, and 8 weeks postimplantation. Histological quantification of total vessel density showed no significant differences between the conditions. Microcomputed tomography indicated significantly higher ectopic bone volume (BV) in the Osteo condition at 4 weeks. At 8 weeks both the Osteo and Blank groups exhibited higher BV compared to the Vasculo and dual Osteo+Vasculo groups. These data not only show that osteogenic microbeads can be used to induce ectopic bone formation, but also suggest an inhibitory effect on BV when undifferentiated bmMSC and HUVEC were included in dual-phase constructs. This work may lead to improved methods for engineering vascularized bone tissue, and to injectable therapies for the treatment of orthopedic pathologies in which bone regeneration is delayed or prevented.


Subject(s)
Bone Development/physiology , Endothelial Cells/cytology , Endothelial Cells/transplantation , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic/physiology , Osteogenesis/physiology , Cell Communication/physiology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques/methods , Endothelial Cells/physiology , Humans , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Tissue Engineering/methods
9.
Acta Biomater ; 10(7): 3091-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24657675

ABSTRACT

Co-culture of endothelial cells (EC) and mesenchymal stem cells (MSC) results in robust vascular network formation in constrained 3-D collagen/fibrin (COL/FIB) composite hydrogels. However, the ability to form endothelial networks is lost when such gels are allowed to compact via cell-mediated remodeling. In this study, we created co-cultures of human EC and human MSC in both constrained and unconstrained COL/FIB matrices and systematically added nanoparticulate hydroxyapatite (HA, 0-20 mg ml(-1)), a bone-like mineral that has been shown to have pro-vasculogenic effects. Constructs cultured for 7 days were assayed for gel compaction, vascular network formation, and mechanical properties. In vitro, robust endothelial network formation was observed in constrained COL/FIB constructs without HA, but this response was significantly inhibited by addition of 5, 10, or 20 mg ml(-1) HA. In unconstrained matrices, network formation was abolished in pure COL/FIB constructs but was rescued by 1.25 or 2.5 mg ml(-1) HA, while higher levels again inhibited vasculogenesis. HA inhibited gel compaction in a dose-dependent manner, which was not correlated to endothelial network formation. HA affected initial stiffness of the gels, but gel remodeling abrogated this effect. Subcutaneous implantation of COL/FIB with 0, 2.5 or 2 0mg ml(-1) HA in the mouse resulted in increased perfusion at the implant site, with no significant differences between materials. Histology at day 7 showed both host and human CD31-stained vasculature infiltrating the implants. These findings are relevant to the design of materials and scaffolds for orthopedic tissue engineering, where both vasculogenesis and formation of a mineral phase are required for regeneration.


Subject(s)
Collagen , Durapatite , Fibrin , Hydrogels , Animals , Cells, Cultured , Coculture Techniques , Humans , In Vitro Techniques , Male , Mice , Mice, SCID
10.
PLoS One ; 9(1): e85749, 2014.
Article in English | MEDLINE | ID: mdl-24465680

ABSTRACT

Non-destructive monitoring of engineered tissues is needed for translation of these products from the lab to the clinic. In this study, non-invasive, high resolution spectral ultrasound imaging (SUSI) was used to monitor the differentiation of MC3T3 pre-osteoblasts seeded within collagen hydrogels. SUSI was used to measure the diameter, concentration and acoustic attenuation of scatterers within such constructs cultured in either control or osteogenic medium over 21 days. Conventional biochemical assays were used on parallel samples to determine DNA content and calcium deposition. Construct volume and morphology were accurately imaged using ultrasound. Cell diameter was estimated to be approximately 12.5-15.5 µm using SUSI, which corresponded well to measurements of fluorescently stained cells. The total number of cells per construct assessed by quantitation of DNA content decreased from 5.6±2.4×10(4) at day 1 to 0.9±0.2×10(4) at day 21. SUSI estimation of the equivalent number of acoustic scatters showed a similar decreasing trend, except at day 21 in the osteogenic samples, which showed a marked increase in both scatterer number and acoustic impedance, suggestive of mineral deposition by the differentiating MC3T3 cells. Estimation of calcium content by SUSI was 41.7±11.4 µg/ml, which agreed well with the biochemical measurement of 38.7±16.7 µg/ml. Color coded maps of parameter values were overlaid on B-mode images to show spatiotemporal changes in cell diameter and calcium deposition. This study demonstrates the use of non-destructive ultrasound imaging to provide quantitative information on the number and differentiated state of cells embedded within 3D engineered constructs, and therefore presents a valuable tool for longitudinal monitoring of engineered tissue development.


Subject(s)
Cell Differentiation , Osteoblasts/cytology , Osteoblasts/diagnostic imaging , Tissue Engineering/methods , 3T3 Cells , Animals , Calcium/metabolism , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Size , Cell Survival , Collagen/metabolism , Mice , Osteoblasts/metabolism , Osteogenesis , Reproducibility of Results , Time Factors , Tissue Engineering/instrumentation , Ultrasonography
11.
J Assoc Physicians India ; 62(11): 50-2, 2014 Nov.
Article in English | MEDLINE | ID: mdl-26281483

ABSTRACT

Congenital mitral stenosis (MS) is a rare congenital cardiac malformation and the obstruction to the flow across the mitral valve can be caused by supramitral ring, commissural fusion, short chordae, anomalous mitral arcade, anomalous position of the papillary muscles and the so-called'parachute mitral valve'. We describe here the case of a 47 year old male diagnosed to have a double outlet right ventricle (DORV), subaortic ventricular septal defect (VSD) with no pulmonary stenosis, severe pulmonary hypertension and congenital MS due to parachute mitral valve.


Subject(s)
Abnormalities, Multiple , Double Outlet Right Ventricle/diagnosis , Hypertension, Pulmonary/etiology , Mitral Valve Stenosis/diagnosis , Mitral Valve/abnormalities , Double Outlet Right Ventricle/complications , Echocardiography , Humans , Hypertension, Pulmonary/diagnosis , Male , Middle Aged , Mitral Valve Stenosis/complications , Mitral Valve Stenosis/congenital , Radiography, Thoracic
12.
Cytotherapy ; 15(11): 1309-22, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23999157

ABSTRACT

This review summarizes recent efforts to create vascularized bone tissue in vitro and in vivo through the use of cell-based therapy approaches. The treatment of large and recalcitrant bone wounds is a serious clinical problem, and in the United States approximately 10% of all fractures are complicated by delayed union or non-union. Treatment approaches with the use of growth factor and gene delivery have shown some promise, but results are variable and clinical complications have arisen. Cell-based therapies offer the potential to recapitulate key components of the bone-healing cascade, which involves concomitant regeneration of vasculature and new bone tissue. For this reason, osteogenic and vasculogenic cell types have been combined in co-cultures to capitalize on the function of each cell type and to promote heterotypic interactions. Experiments in both two-dimensional and three-dimensional systems have provided insight into the mechanisms by which osteogenic and vasculogenic cells interact to form vascularized bone, and these approaches have been translated to ectopic and orthotopic models in small-animal studies. The knowledge generated by these studies will inform and facilitate the next generation of pre-clinical studies, which are needed to move cell-based orthopaedic repair strategies into the clinic. The science and application of cytotherapy for repair of large and ischemic bone defects is developing rapidly and promises to provide new treatment methods for these challenging clinical problems.


Subject(s)
Bone Transplantation/methods , Bone and Bones/blood supply , Cell- and Tissue-Based Therapy/methods , Neovascularization, Physiologic/drug effects , Bone Regeneration , Bone and Bones/injuries , Bone and Bones/surgery , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Humans , Osteoblasts/physiology , Osteogenesis/physiology , Tissue Engineering/methods
13.
J Biomed Mater Res A ; 101(6): 1531-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23554144

ABSTRACT

Modular tissue engineering applies biomaterials-based approaches to create discrete cell-seeded microenvironments, which can be further assembled into larger constructs for the repair of injured tissues. In the current study, we embedded human bone marrow-derived mesenchymal stem cells (MSC) and human adipose-derived stem cells (ASC) in collagen/fibrin (COL/FIB) and collagen/fibrin/hydroxyapatite (COL/FIB/HA) microbeads, and evaluated their suitability for bone tissue engineering applications. Microbeads were fabricated using a water-in-oil emulsification process, resulting in an average microbead diameter of approximately 130 ± 25 µm. Microbeads supported both cell viability and cell spreading of MSC and ASC over 7 days in culture. The embedded cells also began to remodel and compact the microbead matrix as demonstrated by confocal reflectance microscopy imaging. After two weeks of culture in media containing osteogenic supplements, both MSC and ASC deposited calcium mineral in COL/FIB microbeads, but not in COL/FIB/HA microbeads. There were no significant differences between MSC and ASC in any of the assays examined, suggesting that either cell type may be an appropriate cell source for orthopedic applications. This study has implications in the creation of defined microenvironments for bone repair, and in developing a modular approach for delivery of pre-differentiated cells.


Subject(s)
Cell Differentiation/drug effects , Ceramics/pharmacology , Collagen/pharmacology , Congresses as Topic , Fibrin/pharmacology , Mesenchymal Stem Cells/cytology , Osteogenesis/drug effects , Adipose Tissue/cytology , Analysis of Variance , Animals , Biocompatible Materials/pharmacology , Bone Marrow Cells/cytology , Boston , Cattle , Cell Survival/drug effects , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Microscopy, Confocal , Microspheres , Particle Size
14.
Cells Tissues Organs ; 197(5): 333-43, 2013.
Article in English | MEDLINE | ID: mdl-23571151

ABSTRACT

Microencapsulation and delivery of stem cells in biomaterials is a promising approach to repairing damaged tissue in a minimally invasive manner. An appropriate biomaterial niche can protect the embedded cells from the challenging environment in the host tissue, while also directing stem cell differentiation toward the desired lineage. In this study, adult human mesenchymal stem cells (MSC) were embedded in hydrogel microbeads consisting of chitosan and type I collagen using an emulsification process. Glyoxal and ß-glycerophosphate were used as chemical and physical crosslinkers to initiate copolymerization of the matrix materials. The average size and size distribution of the microbeads could be varied by controlling the emulsification conditions. Spheroidal microbeads ranging in diameter from 82 ± 19 to 290 ± 78 µm were produced. Viability staining showed that MSC survived the encapsulation process (>90% viability) and spread inside the matrix over a period of 9 days in culture. Induced osteogenic differentiation using medium supplements showed that MSC increased gene expression of osterix and osteocalcin over time in culture, and also deposited calcium mineral. Bone sialoprotein and type I collagen gene expression were not affected. Delivery of microbeads through standard needles at practically relevant flow rates did not adversely affect cell viability, and microbeads could also be easily molded into prescribed geometries for delivery. Such protein-based microbeads may have utility in orthopedic tissue regeneration by allowing minimally invasive delivery of progenitor cells in microenvironments that are both protective and instructive.


Subject(s)
Bone and Bones/pathology , Chitosan/pharmacology , Collagen/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Microspheres , Wound Healing/drug effects , Adult , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Animals , Bone and Bones/drug effects , Calcium/metabolism , Cattle , Cell Differentiation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Humans , Injections , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Particle Size
15.
Tissue Eng Part C Methods ; 19(7): 507-17, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23134479

ABSTRACT

Biomineral coatings have been extensively used to enhance the osteoconductivity of polymeric scaffolds. Numerous porous scaffolds have previously been coated with a bone-like apatite mineral through incubation in simulated body fluid (SBF). However, characterization of the mineral layer formed on scaffolds, including the amount of mineral within the scaffolds, often requires destructive methods. We have developed a method using micro-computed tomography (µ-CT) scanning to nondestructively quantify the amount of mineral in vitro and in vivo on biodegradable scaffolds made of poly (L-lactic acid) (PLLA) and poly (ε-caprolactone) (PCL). PLLA and PCL scaffolds were fabricated using an indirect solid freeform fabrication (SFF) technique to achieve orthogonally interconnected pore architectures. Biomineral coatings were formed on the fabricated PLLA and PCL scaffolds after incubation in modified SBF (mSBF). Scanning electron microscopy and X-ray diffraction confirmed the formation of an apatite-like mineral. The scaffolds were implanted into mouse ectopic sites for 3 and 10 weeks. The presence of a biomineral coating within the porous scaffolds was confirmed through plastic embedding and µ-CT techniques. Tissue mineral content (TMC) and volume of mineral on the scaffold surfaces detected by µ-CT had a strong correlation with the amount of calcium measured by the orthocresolphthalein complex-one (OCPC) method before and after implantation. There was a strong correlation between OCPC pre- and postimplantation and µ-CT measured TMC (R(2)=0.96 preimplant; R(2)=0.90 postimplant) and mineral volume (R(2)=0.96 preimplant; R(2)=0.89 postimplant). The µ-CT technique showed increases in mineral following implantation, suggesting that µ-CT can be used to nondestructively determine the amount of calcium on coated scaffolds.


Subject(s)
Coated Materials, Biocompatible/pharmacology , Lactic Acid/pharmacology , Minerals/pharmacology , Polyesters/pharmacology , Polymers/pharmacology , Tissue Scaffolds/chemistry , X-Ray Microtomography/methods , Animals , Calcium/pharmacology , Crystallization , Humans , Implants, Experimental , Mice , Microscopy, Electron, Scanning , Spectrometry, X-Ray Emission , X-Ray Diffraction
16.
Adv Healthc Mater ; 2(5): 673-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23184758

ABSTRACT

Modular assembly of protein-polysaccharide microenvironments into 3D macroscale tissue constructs is reported. Rapid and simple centrifugation and vacuum molding methods are used to create cohesive multiphase constructs with prescribed geometries. Human fibroblasts are shown to survive in the microenvironments and in the macroscale constructs. Control of the spatial organization in engineered tissues is a key to recreating the complex tissue architectures needed for regenerative therapies.


Subject(s)
Biocompatible Materials/chemical synthesis , Biomimetic Materials/chemistry , Chitosan/chemistry , Collagen Type I/chemistry , Fibroblasts/cytology , Fibroblasts/physiology , Tissue Engineering/methods , Cell Proliferation , Cell Survival , Cells, Cultured , Cellular Microenvironment , Extracellular Matrix/chemistry , Humans , Materials Testing
17.
Tissue Eng Part C Methods ; 18(12): 935-46, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22624791

ABSTRACT

As tissue engineering products move toward the clinic, nondestructive methods to monitor their development and ensure quality are needed. In this study, high-resolution spectral ultrasound imaging (SUSI) was used to noninvasively characterize mineral content in collagen hydrogels. SUSI was used to generate three-dimensional (3D) grayscale (GS) images of construct morphology with submillimeter resolution. Spectral analysis of the backscattered radio frequency (RF) ultrasound signals was used to determine the midband fit (MBF) and slope of the linearized RF spectrum. These parameters are operator and instrument independent, and were used to characterize the spatial distribution of mineral in constructs supplemented with hydroxyapatite particles. GS and MBF correlated closely with mineral content, while slope was not dependent on concentration. SUSI also was used to monitor mineralization of collagen constructs by immersion in simulated body fluid (SBF) over 21 days. The construct surface was mineralized before the interior, and there was a dose-dependent effect of SBF concentration on degree of mineralization and deposited particle size. MBF density was closely correlated with the amount of calcium deposited. These data demonstrate that SUSI has utility as a noninvasive imaging method for quantitative analysis of mineralization in 3D protein constructs. Such techniques may assist the development of engineered orthopedic tissues.


Subject(s)
Collagen/chemistry , Hydrogels , Ultrasonics , Body Fluids , Calcium/chemistry
18.
Angiogenesis ; 15(2): 253-64, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22382584

ABSTRACT

Co-cultures of endothelial cells (EC) and mesenchymal stem cells (MSC) in three-dimensional (3D) protein hydrogels can be used to recapitulate aspects of vasculogenesis in vitro. MSC provide paracrine signals that stimulate EC to form vessel-like structures, which mature as the MSC transition to the role of mural cells. In this study, vessel-like network formation was studied using 3D collagen/fibrin (COL/FIB) matrices seeded with embedded EC and MSC and cultured for 7 days. The EC:MSC ratio was varied from 5:1, 3:2, 1:1, 2:3 and 1:5. The matrix composition was varied at COL/FIB compositions of 100/0 (pure COL), 60/40, 50/50, 40/60 and 0/100 (pure FIB). Vasculogenesis was markedly decreased in the highest EC:MSC ratio, relative to the other cell ratios. Network formation increased with increasing fibrin content in composite materials, although the 40/60 COL/FIB and pure fibrin materials exhibited the same degree of vasculogenesis. EC and MSC were co-localized in vessel-like structures after 7 days and total cell number increased by approximately 70%. Mechanical property measurements showed an inverse correlation between matrix stiffness and network formation. The effect of matrix stiffness was further investigated using gels made with varying total protein content and by crosslinking the matrix using the dialdehyde glyoxal. This systematic series of studies demonstrates that matrix composition regulates vasculogenesis in 3D protein hydrogels, and further suggests that this effect may be caused by matrix mechanical properties. These findings have relevance to the study of neovessel formation and the development of strategies to promote vascularization in transplanted tissues.


Subject(s)
Collagen/chemistry , Endothelial Cells/cytology , Extracellular Matrix/chemistry , Fibrin/chemistry , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic , Cells, Cultured , Coculture Techniques , Endothelial Cells/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Time Factors
19.
Acta Biomater ; 8(4): 1560-5, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22266029

ABSTRACT

Induced biomineralization of materials has been employed as a strategy to increase integration with host tissue, and more recently as a method to control cell function in tissue engineering. However, mineralization is typically performed in the absence of cells, since hypertonic solutions that lack the nutrients and culture components required for the maintenance of cell viability are often used. In the present study, we exposed fibroblast-seeded three-dimensional collagen-chitosan hydrogels to a defined culture medium modified to have specific concentrations of ions involved in biomineralization. The modified medium caused a significant increase in calcium deposition in collagen-chitosan gels, relative to constructs incubated in a standard medium, though serum supplementation attenuated mineral deposition. Collagen-chitosan constructs became opaque over 3 days of mineralization in modified Dulbecco's modified Eagle medium (DMEM), in contrast to translucent control gels incubated in standard DMEM. Histological staining confirmed increased levels of mineral in the treated constructs. Rheological characterization showed that both the storage and loss moduli increased significantly in mineralized materials. Mineralization of fibroblast-seeded constructs resulted in decreased cell viability and proliferation rate over 3 days of incubation in modified medium, but the cell population remained over 75% viable and regained its proliferative potential after rescue in standard culture medium. The ability to mineralize protein matrices in the presence of cells could be useful in creating mechanically stable tissue constructs, as well as to study the effects of the tissue microenvironment on cell function.


Subject(s)
Chitosan/pharmacology , Collagen/pharmacology , Culture Media/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Hydrogels/pharmacology , Minerals/metabolism , Animals , Calcium/metabolism , Cattle , Cell Survival/drug effects , DNA/metabolism , Fibroblasts/metabolism , Humans , Ions , Microscopy, Confocal , Serum/metabolism
20.
Cell Tissue Res ; 340(1): 81-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20165885

ABSTRACT

Cellular signaling via epidermal growth factor (EGF) and EGF-like ligands can determine cell fate and behavior. Osteoblasts, which are responsible for forming and mineralizing osteoid, express EGF receptors and alter rates of proliferation and differentiation in response to EGF receptor activation. Transgenic mice over-expressing the EGF-like ligand betacellulin (BTC) exhibit increased cortical bone deposition; however, because the transgene is ubiquitously expressed in these mice, the identity of cells affected by BTC and responsible for increased cortical bone thickness remains unknown. We have therefore examined the influence of BTC upon mesenchymal stem cell (MSC) and pre-osteoblast differentiation and proliferation. BTC decreases the expression of osteogenic markers in both MSCs and pre-osteoblasts; interestingly, increases in proliferation require hypoxia-inducible factor-alpha (HIF-alpha), as an HIF antagonist prevents BTC-driven proliferation. Both MSCs and pre-osteoblasts express EGF receptors ErbB1, ErbB2, and ErbB3, with no change in expression under osteogenic differentiation. These are the first data that demonstrate an influence of BTC upon MSCs and the first to implicate HIF-alpha in BTC-mediated proliferation.


Subject(s)
Cell Differentiation/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Osteogenesis/genetics , Adolescent , Betacellulin , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...