Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Hum Pathol ; 67: 205-210, 2017 09.
Article in English | MEDLINE | ID: mdl-28843711

ABSTRACT

Sox10 labeling by immunohistochemistry has been primarily reported in tumors of neural crest origin, such as nerve sheath tumors and melanoma. However, Sox10 also labels primary breast carcinomas, particularly those with the basal-like, triple-negative phenotype. However, the utility of Sox10 labeling in metastatic breast carcinomas has not been reported. Here, we prospectively evaluated Sox10 labeling in surgically resected metastatic breast carcinomas from 26 patients sampled on tissue microarrays. In this cohort, Sox10 labeling was seen in 3 metastatic breast carcinomas (12%), all of which were grade III, triple-negative ductal carcinomas metastatic to the brain (n=2) or lung (n=1). Overall, 38% of triple-negative metastases were Sox10 positive, compared to 0% of estrogen receptor (ER)+ or human epidermal growth factor 2 (HER-2) + metastases (P=.045). In addition, we retrospectively reviewed the use of Sox10 immunohistochemistry in metastatic carcinomas in our clinical practice. We identified 21 cases from January 2012-July 2017 in which Sox10 immunohistochemistry was ordered on clinical sign-out in the work-up of a metastatic carcinoma as being of possible breast origin. Overall, Sox10 labeled 57% (n=12) of all evaluated metastatic carcinomas. All of the Sox10+ tumors were ER-, such that 71% of ER- carcinomas were Sox10+ in comparison to 0% of ER+ carcinomas (P=0.049). In conclusion, the differential diagnosis of a Sox10+ malignancy of unknown origin should not be limited to metastatic melanoma. Sox10 labeling is seen in a subset of metastatic triple-negative breast carcinomas, supporting its use as a marker of breast origin in this setting.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Carcinoma, Ductal, Breast/chemistry , Neoplasms, Unknown Primary/chemistry , SOXE Transcription Factors/analysis , Triple Negative Breast Neoplasms/chemistry , Adult , Aged , Baltimore , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/surgery , Female , Humans , Immunohistochemistry , Lung Neoplasms/secondary , Middle Aged , Neoplasm Grading , Neoplasms, Unknown Primary/pathology , Neoplasms, Unknown Primary/surgery , Predictive Value of Tests , Prospective Studies , Receptors, Estrogen/analysis , Retrospective Studies , Tissue Array Analysis , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/surgery
2.
Plast Reconstr Surg ; 135(5): 1489-1498, 2015 May.
Article in English | MEDLINE | ID: mdl-25919260

ABSTRACT

BACKGROUND: The surgical implantation of materials and devices has dramatically increased over the past decade. This trend is expected to continue with the broadening application of biomaterials and rapid expansion of aging populations. One major factor that limits the potential of implantable materials and devices is the foreign body response, an immunologic reaction characterized by chronic inflammation, foreign body giant cell formation, and fibrotic capsule formation. METHODS: The English literature on the foreign body response to implanted materials and devices is reviewed. RESULTS: Fibrotic encapsulation can cause device malfunction and dramatically limit the function of an implanted medical device or material. Basic science studies suggest a role for immune and inflammatory pathways at the implant-host interface that drive the foreign body response. Current strategies that aim to modulate the host response and improve construct biocompatibility appear promising. CONCLUSIONS: This review article summarizes recent basic science, preclinical, and clinicopathologic studies examining the mechanisms driving the foreign body response, with particular focus on breast implants and synthetic meshes. Understanding these molecular and cellular mechanisms will be critical for achieving the full potential of implanted biomaterials to restore human tissues and organs.


Subject(s)
Biocompatible Materials/adverse effects , Bioengineering , Foreign-Body Reaction/prevention & control , Plastic Surgery Procedures/standards , Prostheses and Implants/adverse effects , Humans
3.
Ann Surg ; 260(6): 1138-46, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25389925

ABSTRACT

OBJECTIVE: To investigate how epithelial mechanotransduction pathways impact wound repair. BACKGROUND: Mechanical forces are increasingly recognized to influence tissue repair, but their role in chronic wound pathophysiology remains unknown. Studies have shown that chronic wounds exhibit high levels of matrix metalloproteinase 9 (MMP9), a key proteolytic enzyme that regulates wound remodeling. We hypothesized that epithelial mechanosensory pathways regulated by keratinocyte-specific focal adhesion kinase (FAK) control dermal remodeling via MMP9. METHODS: A standard wound model was applied to keratinocyte-specific FAK knockout (KO) and control mice. Rates of wound healing were measured and tissue was obtained for histologic and molecular analyses. Transcriptional and immunoblot assays were used to assess the activation of FAK, intracellular kinases, and MMP9 in vitro. A cell suspension model was designed to validate the importance of FAK mechanosensing, p38, and MMP9 secretion in human cells. Biomechanical testing was utilized to evaluate matrix tensile properties in FAK KO and control wounds. RESULTS: Wound healing in FAK KO mice was significantly delayed compared with controls (closure at 15 days compared with 20 days, P = 0.0003). FAK KO wounds demonstrated decreased dermal thickness and collagen density. FAK KO keratinocytes exhibited overactive p38 and MMP9 signaling in vitro, findings recapitulated in human keratinocytes via the deactivation of FAK in the cell suspension model. Functionally, FAK KO wounds were significantly weaker and more brittle than control wounds, results consistent with the histologic and molecular analyses. CONCLUSIONS: Keratinocyte FAK is highly responsive to mechanical cues and may play a critical role in matrix remodeling via regulation of p38 and MMP9. These findings suggest that aberrant epithelial mechanosensory pathways may contribute to pathologic dermal proteolysis and wound chronicity.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/genetics , Keratinocytes/ultrastructure , RNA/genetics , Skin/injuries , Up-Regulation , Wound Healing , Wounds and Injuries/genetics , Animals , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Focal Adhesion Protein-Tyrosine Kinases/biosynthesis , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Infant, Newborn , Keratinocytes/metabolism , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Proteolysis , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
4.
Plast Reconstr Surg ; 132(3): 580-589, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23985633

ABSTRACT

BACKGROUND: Many breast cancer patients are plagued by the disabling complication of upper limb lymphedema after axillary surgery. Conservative treatments using massage and compression therapy do not offer a lasting relief, as they fail to address the chronic transformation of edema into excess adipose tissue. Liposuction to address the adipose nature of the lymphedema has provided an opportunity for a detailed analysis of the stromal fraction of lymphedema-associated fat to clarify the molecular mechanisms for this adipogenic transformation. METHODS: Adipose-derived stem cells were harvested from human lipoaspirate of the upper extremity from age-matched patients with lymphedema (n = 3) or subcutaneous adipose tissue from control patients undergoing cosmetic procedures (n = 3). Immediately after harvest, adipose-derived stem cells were analyzed using single-cell transcriptional profiling techniques. Osteogenic, adipogenic, and vasculogenic gene expression and differentiation were assessed by quantitative real-time polymerase chain reaction and standard in vitro differentiation assays. RESULTS: Differential transcriptional clusters of adipose-derived stem cells were found between lymphedema and subcutaneous fat. Interestingly, lymphedema-associated stem cells had a much higher adipogenic gene expression and enhanced ability to undergo adipogenic differentiation. Conversely, they had lower vasculogenic gene expression and diminished capability to form tubules in vitro, whereas the osteogenic differentiation capacity was not significantly altered. CONCLUSIONS: Adipose-derived stem cells from extremities affected by lymphedema appear to exhibit transcriptional profiles similar to those of abdominal adipose-derived stem cells; however, their adipogenic differentiation potential is strongly increased and their vasculogenic capacity is compromised. These results suggest that the underlying pathophysiology of lymphedema drives adipose-derived stem cells toward adipogenic differentiation.


Subject(s)
Adipogenesis/genetics , Lymphedema/physiopathology , Postoperative Complications/physiopathology , Stem Cells/physiology , Subcutaneous Fat/physiopathology , Transcriptome , Adult , Arm , Axilla , Breast Neoplasms/surgery , Case-Control Studies , Female , Gene Expression Profiling , Genetic Markers , Humans , Lipectomy , Lymph Node Excision , Lymphedema/etiology , Lymphedema/genetics , Lymphedema/surgery , Middle Aged , Postoperative Complications/genetics , Postoperative Complications/surgery , Real-Time Polymerase Chain Reaction
5.
J Burn Care Res ; 33(4): 463-70, 2012.
Article in English | MEDLINE | ID: mdl-22683987

ABSTRACT

Heterotopic ossification (HO), or the abnormal development of bone tissue in soft-tissue locations, can be physically debilitating and clinically devastating. For unclear reasons, HO is highly associated with burn injury. The objective of this review is to summarize 1) cells that are responsible for HO, 2) in vitro and in vivo models of HO and how they have contributed to our current knowledge of the disease process, 3) the effects of the adipose compartment on HO, 4) the effects of inflammation on HO, and 5) the effects of mesenchymal stem cells (MSCs) on HO. Preclinical models of HO suggest several possible mechanisms for the development of this pathologic process, including progenitor cell differentiation and paracrine modulation of local inflammatory responses. Further studies are needed to elucidate the molecular mechanisms driving HO so that targeted therapies can be developed. Current literature supports a role for MSCs in modulating heterotopic bone formation, and direct manipulation of MSCs might one day be used to prevent and treat HO.


Subject(s)
Burns/complications , Burns/therapy , Mesenchymal Stem Cell Transplantation/methods , Ossification, Heterotopic/etiology , Ossification, Heterotopic/therapy , Animals , Burns/diagnosis , Disease Models, Animal , Humans , In Vitro Techniques , Injury Severity Score , Mice , Mice, Transgenic , Ossification, Heterotopic/physiopathology , Prognosis , Rabbits , Risk Assessment , Subcutaneous Fat/physiopathology , Treatment Outcome
7.
Plast Reconstr Surg ; 129(1): 53-66, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21915082

ABSTRACT

BACKGROUND: Previous studies have demonstrated the role of noggin, a bone morphogenetic protein-2 inhibitor, in vascular development and angiogenesis. The authors hypothesized that noggin suppression in human adipose-derived stromal cells would enhance vascular endothelial growth factor secretion and angiogenesis in vitro and in vivo to a greater extent than bone morphogenetic protein-2 alone. METHODS: Human adipose-derived stromal cells were isolated from human lipoaspirate (n = 6) noggin was knocked down using lentiviral techniques. Knockdown was confirmed and angiogenesis was assessed by tubule formation and quantitative real-time polymerase chain reaction. Cells were seeded onto scaffolds and implanted into a 4-mm critical size calvarial defect. In vivo angiogenic signaling was assessed by immunofluorescence and immunohistochemistry. RESULTS: Human adipose-derived stromal cells with noggin suppression secreted significantly higher amounts of angiogenic proteins, expressed higher levels of angiogenic genes, and formed more tubules in vitro. In vivo, calvarial defects seeded with noggin shRNA human adipose-derived stromal cells exhibited a significantly higher number of vessels in the defect site than controls by immunohistochemistry (p < 0.05). In addition, bone morphogenetic protein-2-releasing scaffolds significantly enhanced vascular signaling in the defect site. CONCLUSIONS: Human adipose-derived stromal cells demonstrate significant increases in angiogenesis in vitro and in vivo with both noggin suppression and BMP-2 supplementation. By creating a cell with noggin suppressed and by using a scaffold with increased bone morphogenetic protein-2 signaling, a more angiogenic niche can be created.


Subject(s)
Bone Morphogenetic Protein 2/antagonists & inhibitors , Carrier Proteins/antagonists & inhibitors , Gene Knockdown Techniques , Neovascularization, Physiologic , Stromal Cells/cytology , Adipose Tissue/cytology , Animals , Blotting, Western , Bone Morphogenetic Protein 2/pharmacology , Immunohistochemistry , Mice , Mice, Nude , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Parietal Bone/injuries , Parietal Bone/pathology , Parietal Bone/physiopathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Stromal Cells/metabolism , Stromal Cells/transplantation , Tissue Scaffolds , Vascular Endothelial Growth Factor A/metabolism , Wound Healing/drug effects , Wound Healing/genetics
8.
Nat Med ; 18(1): 148-52, 2011 Dec 11.
Article in English | MEDLINE | ID: mdl-22157678

ABSTRACT

Exuberant fibroproliferation is a common complication after injury for reasons that are not well understood. One key component of wound repair that is often overlooked is mechanical force, which regulates cell-matrix interactions through intracellular focal adhesion components, including focal adhesion kinase (FAK). Here we report that FAK is activated after cutaneous injury and that this process is potentiated by mechanical loading. Fibroblast-specific FAK knockout mice have substantially less inflammation and fibrosis than control mice in a model of hypertrophic scar formation. We show that FAK acts through extracellular-related kinase (ERK) to mechanically trigger the secretion of monocyte chemoattractant protein-1 (MCP-1, also known as CCL2), a potent chemokine that is linked to human fibrotic disorders. Similarly, MCP-1 knockout mice form minimal scars, indicating that inflammatory chemokine pathways are a major mechanism by which FAK mechanotransduction induces fibrosis. Small-molecule inhibition of FAK blocks these effects in human cells and reduces scar formation in vivo through attenuated MCP-1 signaling and inflammatory cell recruitment. These findings collectively indicate that physical force regulates fibrosis through inflammatory FAK-ERK-MCP-1 pathways and that molecular strategies targeting FAK can effectively uncouple mechanical force from pathologic scar formation.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mechanotransduction, Cellular , Skin/enzymology , Skin/pathology , Animals , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cicatrix, Hypertrophic/enzymology , Cicatrix, Hypertrophic/pathology , Fibroblasts/metabolism , Fibrosis , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gene Expression , Humans , MAP Kinase Signaling System , Mice , Mice, Knockout , Mice, Transgenic , Signal Transduction
9.
PLoS One ; 6(10): e25847, 2011.
Article in English | MEDLINE | ID: mdl-22022457

ABSTRACT

INTRODUCTION: The function of Glycogen Synthase Kinases 3ß (GSK-3ß) has previously been shown to be necessary for normal secondary palate development. Using GSK-3ß null mouse embryos, we examine the potential coordinate roles of Wnt and Hedgehog signaling on palatal ossification. METHODS: Palates were harvested from GSK-3ß, embryonic days 15.0-18.5 (e15.0-e18.5), and e15.5 Indian Hedgehog (Ihh) null embryos, and their wild-type littermates. The phenotype of GSK-3ß null embryos was analyzed with skeletal whole mount and pentachrome stains. Spatiotemporal regulation of osteogenic gene expression, in addition to Wnt and Hedgehog signaling activity, were examined in vivo on GSK-3ß and Ihh +/+ and -/- e15.5 embryos using in situ hybridization and immunohistochemistry. To corroborate these results, expression of the same molecular targets were assessed by qRT-PCR of e15.5 palates, or e13.5 palate cultures treated with both Wnt and Hedgehog agonists and anatagonists. RESULTS: GSK-3ß null embryos displayed a 48 percent decrease (*p<0.05) in palatine bone formation compared to wild-type littermates. GSK-3ß null embryos also exhibited decreased osteogenic gene expression that was associated with increased Wnt and decreased Hedgehog signaling. e13.5 palate culture studies demonstrated that Wnt signaling negatively regulates both osteogenic gene expression and Hedgehog signaling activity, while inhibition of Wnt signaling augments both osteogenic gene expression and Hedgehog signaling activity. In addition, no differences in Wnt signaling activity were noted in Ihh null embryos, suggesting that canonical Wnt may be upstream of Hedgehog in secondary palate development. Lastly, we found that GSK-3ß -/- palate cultures were "rescued" with the Wnt inhibitor, Dkk-1. CONCLUSIONS: Here, we identify a critical role for GSK-3ß in palatogenesis through its direct regulation of canonical Wnt signaling. These findings shed light on critical developmental pathways involved in palatogenesis and may lead to novel molecular targets to prevent cleft palate formation.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Mesoderm/embryology , Mesoderm/enzymology , Organogenesis , Osteogenesis , Palate/embryology , Palate/enzymology , Animals , Embryo, Mammalian/enzymology , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3/deficiency , Glycogen Synthase Kinase 3 beta , Hedgehog Proteins/metabolism , Mice , Mice, Inbred C57BL , Mutation/genetics , Organogenesis/genetics , Osteogenesis/genetics , Phenotype , Up-Regulation , Wnt Signaling Pathway
11.
Stem Cells ; 29(12): 2018-29, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21997852

ABSTRACT

An urgent need exists in clinical medicine for suitable alternatives to available techniques for bone tissue repair. Human adipose-derived stem cells (hASCs) represent a readily available, autogenous cell source with well-documented in vivo osteogenic potential. In this article, we manipulated Noggin expression levels in hASCs using lentiviral and nonintegrating minicircle short hairpin ribonucleic acid (shRNA) methodologies in vitro and in vivo to enhance hASC osteogenesis. Human ASCs with Noggin knockdown showed significantly increased bone morphogenetic protein (BMP) signaling and osteogenic differentiation both in vitro and in vivo, and when placed onto a BMP-releasing scaffold embedded with lentiviral Noggin shRNA particles, hASCs more rapidly healed mouse calvarial defects. This study therefore suggests that genetic targeting of hASCs combined with custom scaffold design can optimize hASCs for skeletal regenerative medicine.


Subject(s)
Adipose Tissue/cytology , Bone Regeneration , Osteogenesis , Stem Cells/metabolism , Tissue Scaffolds/chemistry , Adipose Tissue/metabolism , Adult , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation , Female , Gene Knockdown Techniques , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Implants, Experimental , Lactic Acid/chemistry , Lactic Acid/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Male , Mice , Mice, Nude , Mice, Transgenic/genetics , Mice, Transgenic/metabolism , Middle Aged , Polyglycolic Acid/chemistry , Polyglycolic Acid/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Skull/metabolism , Stem Cells/cytology , Time Factors , Tissue Engineering/methods , Young Adult
12.
J Biol Chem ; 286(45): 39497-509, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21949130

ABSTRACT

Clinically available sources of bone for repair and reconstruction are limited by the accessibility of autologous grafts, infectious risks of cadaveric materials, and durability of synthetic substitutes. Cell-based approaches for skeletal regeneration can potentially fill this need, and adipose tissue represents a promising source for development of such therapies. Here, we enriched for an osteogenic subpopulation of cells derived from human subcutaneous adipose tissue utilizing microfluidic-based single cell transcriptional analysis and fluorescence-activated cell sorting (FACS). Statistical analysis of single cell transcriptional profiles demonstrated that low expression of endoglin (CD105) correlated with a subgroup of adipose-derived cells with increased osteogenic gene expression. FACS-sorted CD105(low) cells demonstrated significantly enhanced in vitro osteogenic differentiation and in vivo bone regeneration when compared with either CD105(high) or unsorted cells. Evaluation of the endoglin pathway suggested that enhanced osteogenesis among CD105(low) adipose-derived cells is likely due to identification of a subpopulation with lower TGF-ß1/Smad2 signaling. These findings thus highlight a potential avenue to promote osteogenesis in adipose-derived mesenchymal cells for skeletal regeneration.


Subject(s)
Adipose Tissue/metabolism , Antigens, CD/metabolism , Gene Expression Regulation/physiology , Osteogenesis/physiology , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta1/metabolism , Adipose Tissue/cytology , Adolescent , Adult , Aged , Bone Regeneration/physiology , Cell Differentiation/physiology , Cells, Cultured , Endoglin , Female , Humans , Male , Microfluidic Analytical Techniques , Middle Aged , Smad2 Protein/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Transcription, Genetic/physiology , Transforming Growth Factor beta1/genetics
13.
Plast Reconstr Surg ; 128(2): 373-386, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21788829

ABSTRACT

BACKGROUND: Given the diversity of species from which adipose-derived stromal cells are derived and studied, the authors set out to delineate the differences in the basic cell biology that may exist across species. Briefly, the authors found that significant differences exist with regard to proliferation and osteogenic potentials of adipose-derived stromal cells across species. METHODS: Adipose-derived stromal cells were derived from human, mouse, and canine sources as previously described. Retinoic acid, insulin-like growth factor-1, and bone morphogenetic protein-2 were added to culture medium; proliferation and osteogenic differentiation were assessed by standardized assays. In vivo methods included seeding 150,000 adipose-derived stromal cells on a biomimetic scaffold and analyzing healing by micro-computed tomography and histology. RESULTS: Adipose-derived stromal cells from all species had the capability to undergo osteogenic differentiation. Canine adipose-derived stromal cells were the most proliferative, whereas human adipose-derived stromal cells were the most osteogenic (p < 0.05). Human cells, however, had the most significant osteogenic response to osteogenic media. Retinoic acid stimulated osteogenesis in mouse and canine cells but not in human adipose-derived stromal cells. Insulin-like growth factor-1 enhanced osteogenesis across all species, most notably in human- and canine-derived cells. CONCLUSIONS: Adipose-derived stromal cells derived from human, mouse, and canine all have the capacity to undergo osteogenic differentiation. Canine adipose-derived stromal cells appear to be the most proliferative, whereas human adipose-derived stromal cells appear to be the most osteogenic. Different cytokines and chemicals can be used to modulate this osteogenic response. These results are promising as attempts are made to optimize tissue-engineered bone using adipose-derived stromal cells.


Subject(s)
Adipocytes/physiology , Cell Differentiation/physiology , Osteoblasts/cytology , Osteogenesis/physiology , Stromal Cells/cytology , Adipocytes/ultrastructure , Animals , Cells, Cultured , Culture Media/chemistry , Dogs , Humans , Mice , Microscopy, Electron, Scanning , Middle Aged , Tissue Engineering/methods , X-Ray Microtomography
14.
Stem Cells ; 29(8): 1241-55, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21656608

ABSTRACT

Human adipose-derived stromal cells (hASCs) have a proven capacity to aid in osseous repair of calvarial defects. However, the bone defect microenvironment necessary for osseous healing is not fully understood. In this study, we postulated that the cell-cell interaction between engrafted ASCs and host dura mater (DM) cells is critical for the healing of calvarial defects. hASCs were engrafted into critical sized calvarial mouse defects. The DM-hASC interaction was manipulated surgically by DM removal or by insertion of a semipermeable or nonpermeable membrane between DM and hASCs. Radiographic, histologic, and gene expression analyses were performed. Next, the hASC-DM interaction is assessed by conditioned media (CM) and coculture assays. Finally, bone morphogenetic protein (BMP) signaling from DM was investigated in vivo using novel BMP-2 and anti-BMP-2/4 slow releasing scaffolds. With intact DM, osseous healing occurs both from host DM and engrafted hASCs. Interference with the DM-hASC interaction dramatically reduced calvarial healing with abrogated BMP-2-Smad-1/5 signaling. Using CM and coculture assays, mouse DM cells stimulated hASC osteogenesis via BMP signaling. Through in vivo manipulation of the BMP-2 pathway, we found that BMP-2 plays an important role in DM stimulation of hASC osteogenesis in the context of calvarial bone healing. BMP-2 supplementation to a defect with disrupted DM allowed for bone formation in a nonhealing defect. DM is an osteogenic cell type that both participates in and stimulates osseous healing in a hASC-engrafted calvarial defect. Furthermore, DM-derived BMP-2 paracrine stimulation appears to play a key role for hASC mediated repair.


Subject(s)
Adipose Tissue/pathology , Adult Stem Cells/pathology , Bone Regeneration , Dura Mater/physiopathology , Skull/pathology , Stromal Cells/pathology , Adult , Adult Stem Cells/transplantation , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 2/physiology , Cell Differentiation , Cell Proliferation , Craniocerebral Trauma/therapy , Drug Implants , Dura Mater/pathology , Female , Humans , Mice , Middle Aged , Stromal Cells/transplantation , Tissue Scaffolds , Transplantation, Heterologous , Wound Healing
15.
Genesis ; 49(10): 784-96, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21557453

ABSTRACT

Much is known regarding the role of Indian hedgehog (Ihh) in endochondral ossification, where Ihh regulates multiple steps of chondrocyte differentiation. The Ihh-/- phenotype is most notable for severely foreshortened limbs and a complete absence of mature osteoblasts. A far less explored phenotype in the Ihh-/- mutant is found in the calvaria, where bones form predominately through intramembranous ossification. We investigated the role of Ihh in calvarial bone ossification, finding that proliferation was largely unaffected. Instead, our results indicate that Ihh is a pro-osteogenic factor that positively regulates intramembranous ossification. We confirmed through histologic and quantitative gene analysis that loss of Ihh results in reduction of cranial bone size and all markers of osteodifferentiation. Moreover, in vitro studies suggest that Ihh loss reduces Bmp expression within the calvaria, an observation that may underlie the Ihh-/- calvarial phenotype. In conjunction with the newly recognized roles of Hedgehog deregulation in craniosynostosis, our study defines Ihh as an important positive regulator of cranial bone ossification.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Hedgehog Proteins/metabolism , Osteogenesis , Signal Transduction , Skull/embryology , Animals , Bone Development/genetics , Bone Morphogenetic Proteins/biosynthesis , Bone Morphogenetic Proteins/genetics , Bone and Bones/embryology , Cell Differentiation , Chondrocytes/metabolism , Hedgehog Proteins/biosynthesis , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism
16.
Plast Reconstr Surg ; 127(3): 1118-1129, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21364415

ABSTRACT

BACKGROUND: Studies have demonstrated that human adipose-derived stromal cells (ASCs) are able to repair acute calvarial injuries. The more clinically relevant repair of an established skeletal defect, however, has not been addressed. The authors sought to determine whether human ASCs could heal chronic (established) calvarial defects. METHODS: Critical-sized (4 mm) mouse parietal defects were created. Human ASCs were engrafted either immediately postoperatively (acute defect) or 8 weeks following defect creation (established defect). Methods of analysis included microcomputer tomography scans, histology, and in situ hybridization. Finally, human ASCs were treated in vitro with platelet-rich plasma to simulate an acute wound environment; proliferation and osteogenic differentiation were assessed (alkaline phosphatase, alizarin red, and quantitative reverse transcriptase polymerase chain reaction). RESULTS: Nearly complete osseous healing was observed when calvarial defects were immediately engrafted with human ASCs. In contrast, when human ASCs were engrafted into established defects, little bone formation occurred. Histological analysis affirmed findings by microcomputer tomography, showing more robust staining for alkaline phosphatase and picrosirius red in an acute than in an established human ASC-engrafted defect. In situ hybridization and quantitative reverse transcriptase polymerase chain reaction showed an increase in bone morphogenetic protein (BMP) expression (BMP-2, BMP-4, and BMP-7) acutely following calvarial defect creation. Finally, in vitro treatment of human ASCs with platelet-rich plasma enhanced osteogenic differentiation and increased BMP-2 expression. CONCLUSIONS: Although human ASCs can be utilized to heal an acute mouse calvarial defect, they do not enhance healing of an established (or chronic) defect. Endogenous BMP signaling activated after injury may explain these differences in healing. Platelet-rich plasma enhances osteogenic differentiation of human ASCs in vitro and may prove a promising therapy for future skeletal tissue engineering efforts.


Subject(s)
Adipose Tissue/cytology , Osteogenesis/physiology , Parietal Bone/surgery , Plastic Surgery Procedures/methods , Skull Fractures/surgery , Stem Cell Transplantation/methods , Stromal Cells/cytology , Animals , Cell Proliferation , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Nude , Middle Aged , Parietal Bone/injuries
17.
Plast Reconstr Surg ; 127(3): 1130-1140, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21364416

ABSTRACT

BACKGROUND: Adipose-derived stromal cells are a multipotent cell type with the ability to undergo osteogenic differentiation. The authors sought to examine whether systemically administered adipose-derived stromal cells would migrate to and heal surgically created defects of the mouse cranial skeleton. METHODS: Mouse adipose-derived stromal cells were harvested from luciferase-positive transgenic mice; human adipose-derived stromal cells were harvested from human lipoaspirate and labeled with luciferase and green fluorescent protein. A 4-mm calvarial defect (critical sized) was made in the mouse parietal bone; skin incisions alone were used as a control (n = 5 per group). Adipose-derived stromal cells were injected intravenously (200,000 cells per animal) and compared with saline injection only. Methods of analyses included micro-computed tomographic scanning, in vivo imaging system detection of luciferase activity, and standard histology. RESULTS: Migration of adipose-derived stromal cells to calvarial defect sites was confirmed by accumulation of luciferase activity and green fluorescent protein stain as early as 4 days and persisting up to 4 weeks. Little activity was observed among control groups. Intravenous administration of either mouse or human adipose-derived stromal cells resulted in histologic evidence of bone formation within the defect site, in comparison with an absence of bone among control defects. By micro-computed tomographic analysis, human but not mouse adipose-derived stromal cells stimulated significant osseous healing. CONCLUSIONS: Intravenously administered adipose-derived stromal cells migrate to sites of calvarial injury. Thereafter, intravenous human adipose-derived stromal cells contribute to bony calvarial repair. Intravenous administration of adipose-derived stromal cells may be an effective delivery method for future efforts in skeletal regeneration.


Subject(s)
Adipose Tissue/cytology , Cell Movement , Osteogenesis/physiology , Parietal Bone/injuries , Stem Cell Transplantation/methods , Stromal Cells/metabolism , Wound Healing/physiology , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Female , Humans , Injections, Intravenous , Mice , Mice, Nude , Middle Aged , Parietal Bone/pathology , Stromal Cells/cytology
18.
Plast Reconstr Surg ; 127(3): 1182-1190, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21364421

ABSTRACT

BACKGROUND: Cleft lip-cleft palate is a common congenital disability and represents a large biomedical burden. Through the use of animal models, the molecular underpinnings of cleft palate are becoming increasingly clear. Indian hedgehog (Ihh) has been shown to be associated with craniofacial development and to be active in the palatine bone. The authors hypothesize that Indian hedgehog activity plays a role in osteogenesis within the secondary palate and that defects in this pathway may inhibit osteogenesis of the secondary palate. METHODS: Palates were isolated from wild-type mice during the period of palate development (embryonic days 9.5 to 17.5). Quantitative real-time polymerase chain reaction was used for detecting gene expression during osteogenic differentiation and cellular differentiation (Shh, Ihh, Ptc1, Gli1, Gli2, Gli3, Runx2, Alp, and Col1a1). Next, palates were analyzed by hematoxylin and eosin, aniline blue, pentachrome, and in situ hybridization to assess osteogenesis of the palatal shelf and expression of hedgehog pathway genes. Finally, the palates of Indian hedgehog-null mice were analyzed to determine the effect of genetic deficiency on palatal development osteogenesis. RESULTS: Increased Indian hedgehog and osteogenic signaling coincided with ossification and fusion of the palate in wild-type mice. This included a fivefold to 150-fold peak in expression of hedgehog elements, including Ihh, at embryonic day 15.5 as compared with embryonic day 9.5. Contrarily, loss of Indian hedgehog by genetic knockout (Ihh-/-) resulted in decreased secondary palate ossification. CONCLUSIONS: The authors' results suggest a role for hedgehog signaling during palatal ossification. The hedgehog pathway is activated during palatal fusion, and deletion of Indian hedgehog leads to diminished ossification of the secondary hard palate.


Subject(s)
Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Osteogenesis/physiology , Palate, Hard/embryology , Pregnancy, Animal , RNA/genetics , Signal Transduction/physiology , Animals , Female , Hedgehog Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Palate, Hard/metabolism , Polymerase Chain Reaction , Pregnancy
19.
Stem Cells Dev ; 20(2): 243-57, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20698749

ABSTRACT

Human adipose-derived stromal cells (hASCs) have the proven capacity to ossify skeletal defects. The mechanisms whereby hASCs stimulate bone repair are not fully understood. In this study, we examined the potential for hASCs to stimulate autogenous repair of a mouse calvarial defect. Immunofluoresence, osteogenic stains, and surface electron microscopy were used to demonstrate osteogenic differentiation of hASCs. hASCs were engrafted into 4 mm calvarial defects in athymic mice using an osteoconductive scaffold. Analysis included microcomputed tomography, histology, in situ hybridization, and quantitative real-time-polymerase chain reaction. Next, the in vitro interaction between hASCs and mouse calvarial osteoblasts (mOBs) was assessed by the conditioned medium and coculture assays. The medium was supplemented with Hedgehog signaling modifiers, including recombinant N-terminal Sonic hedgehog, smoothened agonist, and cyclopamine. Finally, cyclopamine was delivered in vivo to hASC-engrafted defects. Significant calvarial healing was observed among hASC-engrafted defects compared with control groups (no treatment or scaffold alone) (*P<0.05). hASCs showed evidence of stimulation of host mouse osteogenesis, including (1) increased expression of bone markers at the defect edge by in situ hybridization, and (2) increased host osteogenic gene expression by species-specific quantitative real-time polymerase chain reaction. Using the conditioned medium or coculture assays, hASCs stimulated mOB osteogenic differentiation, accompanied by Hedgehog signaling activation. N-terminal Sonic hedgehog or smoothened agonist replicated, while cyclopamine reversed, the pro-osteogenic effect of the conditioned medium on mOBs. Finally, cyclopamine injection arrested bone formation in vivo. hASCs heal critical-sized mouse calvarial defects, this is, at least in part, via stimulation of autogenous healing of the host defect. Our studies suggest that hASC-derived Hedgehog signaling may play a paracrine role in skeletal repair.


Subject(s)
Adipose Tissue/cytology , Bone Regeneration , Hedgehog Proteins/metabolism , Osteoblasts/pathology , Osteogenesis , Paracrine Communication , Skull/pathology , Adipose Tissue/metabolism , Adult , Animals , Cell Differentiation , Cell Transplantation , Cells, Cultured , Coculture Techniques , Female , Gene Expression Profiling , Humans , Implants, Experimental , Male , Mice , Mice, Nude , Mice, Transgenic , Middle Aged , Osteoblasts/metabolism , Signal Transduction , Skull/metabolism , Skull/physiopathology , Stromal Cells/cytology , Stromal Cells/metabolism , Stromal Cells/transplantation , Tissue Scaffolds
20.
Stem Cells Dev ; 20(3): 427-39, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20536327

ABSTRACT

Human adipose-derived stromal cells (hASCs) represent a multipotent stromal cell type with a proven capacity to undergo osteogenic differentiation. Many hurdles exist, however, between current knowledge of hASC osteogenesis and their potential future use in skeletal tissue regeneration. The impact of frozen storage on hASC osteogenic differentiation, for example, has not been studied in detail. To examine the effects of frozen storage, hASCs were harvested from lipoaspirate and either maintained in standard culture conditions or frozen for 2 weeks under standard conditions (90% fetal bovine serum, 10% dimethyl sulfoxide). Next, in vitro parameters of cell morphology (surface electron microscopy [EM]), cell viability and growth (trypan blue; bromodeoxyuridine incorporation), osteogenic differentiation (alkaline phosphatase, alizarin red, and quantitative real-time (RT)-polymerase chain reaction), and adipogenic differentiation (Oil red O staining and quantitative RT-polymerase chain reaction) were performed. Finally, in vivo bone formation was assessed using a critical-sized cranial defect in athymic mice, utilizing a hydroxyapatite (HA)-poly(lactic-co-glycolic acid) scaffold for ASC delivery. Healing was assessed by serial microcomputed tomography scans and histology. Freshly derived ASCs differed significantly from freeze-thaw ASCs in all markers examined. Surface EM showed distinct differences in cellular morphology. Proliferation, and osteogenic and adipogenic differentiation were all significantly hampered by the freeze-thaw process in vitro (*P < 0.01). In vivo, near complete healing was observed among calvarial defects engrafted with fresh hASCs. This was in comparison to groups engrafted with freeze-thaw hASCs that showed little healing (*P < 0.01). Finally, recombinant insulin-like growth factor 1 or recombinant bone morphogenetic protein 4 was observed to increase or rescue in vitro osteogenic differentiation among frozen hASCs (*P < 0.01). The freezing of ASCs for storage significantly impacts their biology, both in vitro and in vivo. The ability of ASCs to successfully undergo osteogenic differentiation after freeze-thaw is substantively muted, both in vitro and in vivo. The use of recombinant proteins, however, may be used to mitigate the deleterious effects of the freeze-thaw process.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation , Cryopreservation , Multipotent Stem Cells/cytology , Alkaline Phosphatase/metabolism , Animals , Bone Regeneration , Cell Adhesion , Cell Proliferation , Cell Survival , Cells, Cultured , Female , Freezing , Gene Expression , Humans , Male , Mice , Mice, Nude , Middle Aged , Multipotent Stem Cells/transplantation , Multipotent Stem Cells/ultrastructure , Osteocytes/cytology , Osteocytes/metabolism , Skull/injuries , Skull/pathology , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...