Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7(1): 13027, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29026147

ABSTRACT

Genome wide studies indicate that vascular endothelial growth factor A (VEGF) is associated with osteoarthritis (OA), and increased VEGF expression correlates with increased disease severity. VEGF is also a chondrocyte survival factor during development and essential for bone formation, skeletal growth and postnatal homeostasis. This raises questions of how the important embryonic and postnatal functions of VEGF can be reconciled with an apparently destructive role in OA. Addressing these questions, we find that VEGF acts as a survival factor in growth plate chondrocytes during development but only up until a few weeks after birth in mice. It is also required for postnatal differentiation of articular chondrocytes and the timely ossification of bones in joint regions. In surgically induced knee OA in mice, a model of post-traumatic OA in humans, increased expression of VEGF is associated with catabolic processes in chondrocytes and synovial cells. Conditional knock-down of Vegf attenuates induced OA. Intra-articular anti-VEGF antibodies suppress OA progression, reduce levels of phosphorylated VEGFR2 in articular chondrocytes and synovial cells and reduce levels of phosphorylated VEGFR1 in dorsal root ganglia. Finally, oral administration of the VEGFR2 kinase inhibitor Vandetanib attenuates OA progression.


Subject(s)
Cartilage, Articular/embryology , Cartilage, Articular/pathology , Osteoarthritis/metabolism , Vascular Endothelial Growth Factor A/metabolism , Administration, Oral , Animals , Antibodies/pharmacology , Bone Development , Cell Differentiation , Cell Lineage , Chondrocytes/metabolism , Collagen Type II/metabolism , Disease Progression , Endothelium/metabolism , Female , Gene Deletion , Gene Expression Regulation , Growth Plate/metabolism , Growth Plate/pathology , Integrases/metabolism , Knee Joint/pathology , Mice, Inbred C57BL , Neovascularization, Physiologic , Osteoarthritis/pathology , Osteogenesis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
2.
Int J Exp Pathol ; 97(4): 296-302, 2016 08.
Article in English | MEDLINE | ID: mdl-27581728

ABSTRACT

Studies of proliferative hemangiomas have led to the discovery that interactions of endothelial cells with extracellular matrix and/or Vascular Endothelial Growth Factor (VEGF)-A stimulate the expression of VEGFR1, the VEGF decoy receptor, and suppress VEGF-dependent VEGFR2 signalling by a mechanism that requires the matrix-binding receptor Anthrax Toxin Receptor (ANTXR)1, VEGFR2, ß1 integrin and the Nuclear Factor of Activated T cells (NFAT). In hemangioma endothelial cells, all these components are present, but are functionally compromised, so that the levels of VEGFR1 are extremely low and VEGFR2 signalling is constitutively active. Consequently, the levels of Hypoxia Inducible Factor (HIF)-1α and its transcriptional targets, VEGF-A and C-X-C motif chemokine 12 (CxCl12), are elevated and a positive VEGF-A feedback loop is established. Overexpression of ANTXR1, carrying a heterozygous Ala-to-Thr mutation, induces hemangioma-like signalling in control endothelial cells; VEGF signalling is normalized when wild-type ANTXR1 is overexpressed in hemangioma cells. These findings suggest that ANTXR1 functions as a negative regulator of VEGF-A signalling. Studies of a mouse model of the Growth Retardation, Alopecia, Pseudo-anodontia and Optic Atrophy (GAPO) syndrome, caused by the loss-of-function mutations in ANTXR1, as well as knock-in mice carrying the Ala-to-Thr ANTXR1 mutation, confirm that ANTXR1 functions as a suppressor of VEGF-A signalling. Cutaneous endothelial cells isolated from ANTXR1-deficient mice exhibit low levels of VEGFR1, elevated levels of VEGF-A, HIF-1α and CxCl12 and activated VEGFR2 signalling as in hemangioma. Increased numbers of myeloid cells in the skin of ANTXR1-deficient mice are associated with reduced vascularity and increased skin fibrosis, suggesting a mechanism for hemangioma involution and replacement by fibrotic scars. Through controlling VEGF-A signalling and extracellular matrix synthesis, ANTXR1 is emerging as a key regulator of skeletal and connective tissue development and homeostasis.


Subject(s)
Bone Development/physiology , Connective Tissue/growth & development , Hemangioma/metabolism , Homeostasis/physiology , Animals , Hemangioma/pathology , Humans , Microfilament Proteins , Neoplasm Proteins/physiology , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
3.
Matrix Biol ; 52-54: 127-140, 2016.
Article in English | MEDLINE | ID: mdl-26899202

ABSTRACT

Deficiency of vascular endothelial growth factor A (VEGF) has been associated with severe craniofacial anomalies in both humans and mice. Cranial neural crest cell (NCC)-derived VEGF regulates proliferation, vascularization and ossification of cartilage and membranous bone. However, the function of VEGF derived from specific subpopulations of NCCs in controlling unique aspects of craniofacial morphogenesis is not clear. In this study a conditional knockdown strategy was used to genetically delete Vegfa expression in Osterix (Osx) and collagen II (Col2)-expressing NCC descendants. No major defects in calvaria and mandibular morphogenesis were observed upon knockdown of VEGF in the Col2(+) cell population. In contrast, loss of VEGF in Osx(+) osteoblast progenitor cells led to reduced ossification of calvarial and mandibular bones without affecting the formation of cartilage templates in newborn mice. The early stages of ossification in the developing jaw revealed decreased initial mineralization levels and a reduced thickness of the collagen I (Col1)-positive bone template upon loss of VEGF in Osx(+) precursors. Increased numbers of proliferating cells were detected within the jaw mesenchyme of mutant embryos. Explant culture assays revealed that mandibular osteogenesis occurred independently of paracrine VEGF action and vascular development. Reduced VEGF expression in mandibles coincided with increased phospho-Smad1/5 (P-Smad1/5) levels and bone morphogenetic protein 2 (Bmp2) expression in the jaw mesenchyme. We conclude that VEGF derived from Osx(+) osteoblast progenitor cells is required for optimal ossification of developing mandibular bones and modulates mechanisms controlling BMP-dependent specification and expansion of the jaw mesenchyme.


Subject(s)
Mandible/growth & development , Neural Crest/cytology , Skull/growth & development , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Animals , Animals, Newborn , Calcification, Physiologic , Cell Proliferation , Cells, Cultured , Collagen Type II/metabolism , Gene Knockdown Techniques , Mandible/metabolism , Mice , Skull/metabolism , Sp7 Transcription Factor/metabolism
4.
Bone ; 80: 14-18, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26453494

ABSTRACT

The development of the vertebrate skeleton reflects its evolutionary history. Cartilage formation came before biomineralization and a head skeleton evolved before the formation of axial and appendicular skeletal structures. This review describes the processes that result in endochondral and intramembranous ossification, the important roles of growth and transcription factors, and the consequences of mutations in some of the genes involved. Following a summary of the origin of cartilage, muscle, and tendon cell lineages in the axial skeleton, we discuss the role of muscle forces in the formation of skeletal architecture and assembly of musculoskeletal functional units. Finally, ontogenetic patterning of bones in response to mechanical loading is reviewed.This article is part of a Special Issue entitled "Muscle Bone Interactions".


Subject(s)
Bone Development/physiology , Animals , Chondrogenesis/physiology , Humans , Osteogenesis/physiology , Tendons/embryology , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Development ; 142(11): 1984-91, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25977369

ABSTRACT

Vascular endothelial growth factor A (Vegfa) has important roles in endochondral bone formation. Osteoblast precursors, endothelial cells and osteoclasts migrate from perichondrium into primary ossification centers of cartilage templates of future bones in response to Vegfa secreted by (pre)hypertrophic chondrocytes. Perichondrial osteolineage cells also produce Vegfa, but its function is not well understood. By deleting Vegfa in osteolineage cells in vivo, we demonstrate that progenitor-derived Vegfa is required for blood vessel recruitment in perichondrium and the differentiation of osteoblast precursors in mice. Conditional deletion of Vegfa receptors indicates that Vegfa-dependent effects on osteoblast differentiation are mediated by Vegf receptor 2 (Vegfr2). In addition, Vegfa/Vegfr2 signaling stimulates the expression and activity of Indian hedgehog, increases the expression of ß-catenin and inhibits Notch2. Our findings identify Vegfa as a regulator of perichondrial vascularity and osteoblast differentiation at early stages of bone development.


Subject(s)
Bone Development , Bone and Bones/blood supply , Cell Differentiation , Neovascularization, Physiologic , Osteoblasts/cytology , Osteoblasts/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Bone and Bones/metabolism , Calcification, Physiologic , Cell Count , Cell Lineage , Hedgehog Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Mice , Osteogenesis , Receptor, Notch2/metabolism , Signal Transduction , Stem Cells/cytology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zinc Finger Protein GLI1 , beta Catenin/metabolism
6.
J Histochem Cytochem ; 62(2): 103-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24309509

ABSTRACT

Vascular endothelial growth factor A (VEGF), a key factor in angiogenesis, plays an essential role in skeletal development and postnatal homeostasis. VEGF serves as a survival factor for chondrocytes and couples the resorption of cartilage with bone formation during endochondral ossification. Recently, it has also been found to regulate the balance between osteoblast and adipocyte differentiation in bone marrow mesenchymal stem cells. Surprisingly, this regulatory function of VEGF is not based on paracrine signaling involving cell surface receptor activation. Instead, the mechanism appears to utilize intracellular VEGF, which is functionally linked to the nuclear envelope protein lamin A. Lamin A and VEGF control osteoblast and adipocyte differentiation by regulating the levels of the osteoblast and adipocyte transcription factors Runx2 and PPARγ, respectively. These data raise the intriguing possibility that loss of bone mass during aging may be manipulated by controlling the levels and activity of intracellular VEGF in bone marrow mesenchymal stem cells.


Subject(s)
Mesenchymal Stem Cells/cytology , Vascular Endothelial Growth Factor A/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Bone and Bones/cytology , Bone and Bones/physiology , Cell Differentiation , Homeostasis , Humans , Mesenchymal Stem Cells/metabolism , Mutation , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Paracrine Communication , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics
7.
Cell Mol Life Sci ; 71(3): 493-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23934155

ABSTRACT

Osteoblasts and adipocytes share a common precursor in adult bone marrow and there is a degree of plasticity between the two cell lineages. This has important implications for the etiology of not only osteoporosis but also several other diseases involving an imbalance between osteoblasts and adipocytes. Understanding the process of differentiation of osteoblasts and adipocytes and their trans-differentiation is crucial in order to identify genes and other factors that may contribute to the pathophysiology of such diseases. Several transcriptional regulators have been shown to control osteoblast and adipocyte differentiation and function. Regulation of cell commitment occurs at the level of the progenitor cell through cross talk between complex signaling pathways and epigenetic mechanisms such as DNA methylation, chromatin remodeling, and microRNAs. Here we review the complex precursor cell microenvironment controlling osteoblastogenesis and adipogenesis during tissue development, maintenance, and pathology.


Subject(s)
Adipocytes/cytology , Bone Marrow Cells/cytology , Cell Differentiation/physiology , Cell Lineage/physiology , Cellular Microenvironment/physiology , Osteoblasts/cytology , Regulatory Elements, Transcriptional/physiology , Animals , Epigenesis, Genetic/physiology , Humans , Regulatory Elements, Transcriptional/genetics
8.
Matrix Biol ; 35: 223-31, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24373744

ABSTRACT

Matrix proteoglycans such as biglycan (Bgn) dominate skeletal tissue and yet its exact role in regulating bone function is still unclear. In this paper we describe the potential role of (Bgn) in the fracture healing process. We hypothesized that Bgn could regulate fracture healing because of previous work showing that it can affect normal bone formation. To test this hypothesis, we created fractures in femurs of 6-week-old male wild type (WT or Bgn+/0) and Bgn-deficient (Bgn-KO or Bgn-/0) mice using a custom-made standardized fracture device, and analyzed the process of healing over time. The formation of a callus around the fracture site was observed at both 7 and 14 days post-fracture in WT and Bgn-deficient mice and immunohistochemistry revealed that Bgn was highly expressed in the fracture callus of WT mice, localizing within woven bone and cartilage. Micro-computed tomography (µCT) analysis of the region surrounding the fracture line showed that the Bgn-deficient mice had a smaller callus than WT mice. Histology of the same region also showed the presence of less cartilage and woven bone in the Bgn-deficient mice compared to WT mice. Picrosirius red staining of the callus visualized under polarized light showed that there was less fibrillar collagen in the Bgn-deficient mice, a finding confirmed by immunohistochemistry using antibodies to type I collagen. Interestingly, real time RT-PCR of the callus at 7 days post-fracture showed a significant decrease in relative vascular endothelial growth factor A (VEGF) gene expression by Bgn-deficient mice as compared to WT. Moreover, VEGF was shown to bind directly to Bgn through a solid-phase binding assay. The inability of Bgn to directly enhance VEGF-induced signaling suggests that Bgn has a unique role in regulating vessel formation, potentially related to VEGF storage or stabilization in the matrix. Taken together, these results suggest that Bgn has a regulatory role in the process of bone formation during fracture healing, and further, that reduced angiogenesis could be the molecular basis.


Subject(s)
Biglycan/metabolism , Fracture Healing/physiology , Neovascularization, Physiologic/physiology , Osteogenesis/physiology , Signal Transduction/physiology , Animals , Bony Callus/diagnostic imaging , Bony Callus/metabolism , DNA Primers/genetics , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , Male , Mice , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/metabolism , X-Ray Microtomography
9.
PLoS One ; 8(8): e71709, 2013.
Article in English | MEDLINE | ID: mdl-23977121

ABSTRACT

Prostate cancer (PC) is a leading cause of death in men however the factors that regulate its progression and eventual metastasis to bone remain unclear. Here we show that WISP1/CCN4 expression in prostate cancer tissues was up-regulated in early stages of the disease and, further, that it correlated with increased circulating levels of WISP1 in the sera of patients at early stages of the disease. WISP1 was also elevated in the mouse prostate cancer model TRAMP in the hypoplastic diseased tissue that develops prior to advanced carcinoma formation. When the ability of anti-WISP1 antibodies to reduce the spread of PC3-Luc cells to distant sites was tested it showed that twice weekly injections of anti-WISP1 antibodies reduced the number and overall size of distant tumors developed after intracardiac (IC) injection of PC3-Luc cells in mice. The ability of antibodies against WISP1 to inhibit growth of PC3-Luc cancer cells in mice was also evaluated and showed that twice weekly injections of anti-WISP1 antibodies reduced local tumor growth when examined in xenografts. To better understand the mechanism of action, the migration of PC3-Luc cells through membranes with or without a Matrigel™ barrier showed the cells were attracted to WISP1, and that this attraction was inhibited by treatment with anti-WISP1 antibodies. We also show the expression of WISP1 at the bone-tumor interface and in the stroma of early grade cancers suggested WISP1 expression is well placed to play roles in both fostering growth of the cancer and its spread to bone. In summary, the up-regulation of WISP1 in the early stages of cancer development coupled with its ability to inhibit spread and growth of prostate cancer cells makes it both a potential target and an accessible diagnostic marker for prostate cancer.


Subject(s)
Bone Neoplasms/secondary , CCN Intercellular Signaling Proteins/metabolism , Molecular Targeted Therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Animals , Antibodies/pharmacology , Bone Neoplasms/pathology , CCN Intercellular Signaling Proteins/blood , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Humans , Immunocompromised Host , Immunohistochemistry , Injections , Luciferases/metabolism , Male , Mice , Neoplasm Invasiveness , Prostatic Neoplasms/blood , Proto-Oncogene Proteins/blood , Xenograft Model Antitumor Assays
10.
J Clin Invest ; 122(9): 3101-13, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22886301

ABSTRACT

Osteoporotic bones have reduced spongy bone mass, altered bone architecture, and increased marrow fat. Bone marrow stem cells from osteoporotic patients are more likely to differentiate into adipocytes than control cells, suggesting that adipocyte differentiation may play a role in osteoporosis. VEGF is highly expressed in osteoblastic precursor cells and is known to stimulate bone formation. Here we tested the hypothesis that VEGF is also an important regulator of cell fate, determining whether differentiation gives rise to osteoblasts or adipocytes. Mice with conditional VEGF deficiency in osteoblastic precursor cells exhibited an osteoporosis-like phenotype characterized by reduced bone mass and increased bone marrow fat. In addition, reduced VEGF expression in mesenchymal stem cells resulted in reduced osteoblast and increased adipocyte differentiation. Osteoblast differentiation was reduced when VEGF receptor 1 or 2 was knocked down but was unaffected by treatment with recombinant VEGF or neutralizing antibodies against VEGF. Our results suggested that VEGF controls differentiation in mesenchymal stem cells by regulating the transcription factors RUNX2 and PPARγ2 as well as through a reciprocal interaction with nuclear envelope proteins lamin A/C. Importantly, our data support a model whereby VEGF regulates differentiation through an intracrine mechanism that is distinct from the role of secreted VEGF and its receptors.


Subject(s)
Adipocytes/physiology , Cell Differentiation , Osteoblasts/physiology , Vascular Endothelial Growth Factor A/deficiency , Adiposity , Alkaline Phosphatase , Animals , Bone Density , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Cells, Cultured , Colony-Forming Units Assay , Core Binding Factor Alpha 1 Subunit/metabolism , Femur/diagnostic imaging , Femur/pathology , Gene Expression , Gene Expression Regulation , Lamin Type A/metabolism , Male , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteogenesis , Osteoporosis/metabolism , Osteoporosis/pathology , PPAR gamma/genetics , PPAR gamma/metabolism , Radiography , Tibia/diagnostic imaging , Tibia/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
J Tissue Eng Regen Med ; 6(9): 721-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-21948715

ABSTRACT

Collagen gels are promising scaffolds to prepare an implant for cartilage repair but several parameters, such as collagen concentration and composition as well as cell density, should be carefully considered, as they are reported to affect phenotypic aspects of chondrocytes. In this study we investigated whether the presence of collagen type I or II in gel lattices affects matrix contraction and relative gene expression levels of matrix proteins, MMPs and the subsequent degradation of collagen by goat articular chondrocytes. Only floating collagen I gels, and not those attached or composed of type II collagen, contracted during a culture period of 12 days. This coincided with an upregulation of both Mmp13 and -14 gene expression, whereas Mmp1 expression was not affected. The release of hydroxyproline in the culture medium, indicating matrix degradation, was increased five-fold in contracted collagen I gels compared to collagen II gels without contraction. Furthermore, blocking contraction of collagen I gels by cytochalasin B inhibited Mmp13 and -14 expression and the release of hydroxyproline. The expression of cartilage-specific ECM genes was decreased in contracted collagen I gels, with increased numbers of cells with an elongated morphology, suggesting that matrix contraction induces dedifferentiation of chondrocytes into fibroblast-like cells. We conclude that the collagen composition of the gels affects matrix contraction by articular chondrocytes and that matrix contraction induces an increased Mmp13 and -14 expression as well as matrix degradation.


Subject(s)
Chondrocytes/metabolism , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 14/genetics , Tissue Engineering/methods , Animals , Cell Count , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Collagen Type I/chemistry , Collagen Type II/chemistry , Cytochalasin B/pharmacology , Extracellular Matrix/metabolism , Gels , Gene Expression/drug effects , Goats , Matrix Metalloproteinase 1/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Scaffolds/chemistry
12.
Cell ; 147(6): 1222-3, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22153068

ABSTRACT

Cherubism, a case of bone remodeling gone haywire, is associated with mutations in the adaptor protein SH3BP2. Two papers in this issue of Cell (Guettler et al. and Levaot et al.) demonstrate that these mutations disrupt the interaction between SH3BP2 and Tankyrase and describe rules for substrate recognition by this poly(ADP-ribose) polymerase. Establishing such rules paves the way to identifying all Tankyrase-regulated pathways in cells.

13.
Proc Natl Acad Sci U S A ; 108(41): 17022-7, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21969569

ABSTRACT

Although extracellular control of canonical Wnt signaling is crucial for tissue homeostasis, the role of the extracellular microenvironment in modulating this signaling pathway is largely unknown. In the present study, we show that a member of the small leucine-rich proteoglycan family, biglycan, enhances canonical Wnt signaling by mediating Wnt function via its core protein. Immunoprecipitation analysis revealed that biglycan interacts with both the canonical Wnt ligand Wnt3a and the Wnt coreceptor low-density lipoprotein receptor-related protein 6 (LRP6), possibly via the formation of a trimeric complex. Biglycan-deficient cells treated with exogenous Wnt3a had less Wnt3a retained in cell layers compared with WT cells. Furthermore, the Wnt-induced levels of LRP6 phosphorylation and expression of several Wnt target genes were blunted in biglycan-deficient cells. Both recombinant biglycan proteoglycan and biglycan core protein increased Wnt-induced ß-catenin/T cell-specific factor-mediated transcriptional activity, and this activity was completely inhibited by Dickkopf 1. Interestingly, recombinant biglycan was able to rescue impaired Wnt signaling caused by a previously described missense mutation in the extracellular domain of human LRP6 (R611C). Furthermore, biglycan's modulation of canonical Wnt signaling affected the functional activities of osteoprogenitor cells, including the RUNX2-mediated transcriptional activity and calcium deposition. Use of a transplant system and a fracture healing model revealed that expression of Wnt-induced secreted protein 1 was decreased in bone formed by biglycan-deficient cells, further suggesting reduced Wnt signaling in vivo. We propose that biglycan may serve as a reservoir for Wnt in the pericellular space and modulate Wnt availability for activation of the canonical Wnt pathway.


Subject(s)
Biglycan/metabolism , Wnt Signaling Pathway/physiology , Animals , Biglycan/deficiency , Biglycan/genetics , Extracellular Matrix/metabolism , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-6/chemistry , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mice , Mice, Knockout , Mutation, Missense , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Skull/metabolism , Wnt3A Protein/genetics , Wnt3A Protein/metabolism , beta Catenin/metabolism
14.
Cells Tissues Organs ; 192(2): 116-24, 2010.
Article in English | MEDLINE | ID: mdl-20185895

ABSTRACT

Stable integration of collagenous tissue-engineered constructs to surrounding solid devices can be accomplished by coating the solid surfaces with exogenous alkaline phosphatase (ALP). We showed previously that coating of culture well surfaces with the enzyme in combination with the presence of its substrate beta-glycerophosphate (beta-GP) induces mineral deposition at the interface of matrix and surface, thereby preventing matrix detachment. In this study the effect of such mineral-inducing conditions on differentiation of human periodontal ligament (PDL) fibroblasts into osteoblasts/cementoblasts was analyzed in three-dimensional collagen gels. Mineral-inducing conditions decreased collagen type I gene expression and induced dentin matrix protein 1 (DMP1; a marker of late osteoblasts/cementoblasts) gene expression by fibroblasts. DMP1 protein was detected in some fibroblasts only in mineralizing gels. Exogenous ALP released high levels of inorganic phosphate from beta-GP. Addition of inorganic phosphate alone induced DMP1 gene expression, which could be prevented by blocking phosphate entry into fibroblasts by foscarnet. We concluded that mineralizing conditions induced by exogenous ALP affect the phenotype of PDL fibroblasts. The fibroblasts are stimulated to express the late osteoblast/osteocyte marker protein DMP1, which is mediated by uptake of inorganic phosphate into the cells. The enzyme-mediated mineral deposition may thus facilitate enhanced integration of collagenous tissue-engineered constructs to devices or implants in vitro.


Subject(s)
Collagen Type I/biosynthesis , Extracellular Matrix Proteins/biosynthesis , Fibroblasts/metabolism , Periodontal Ligament/metabolism , Phosphoproteins/biosynthesis , Adult , Cell Differentiation/drug effects , Collagen , Glycerophosphates/pharmacology , Humans , Male , Osteopontin/biosynthesis , Periodontal Ligament/cytology , Phosphates/pharmacology
15.
Tissue Eng Part C Methods ; 15(4): 561-70, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19216643

ABSTRACT

In this study we present a new three-dimensional (3D) model to study effects of mechanical loading on tendon/ligament formation in vitro. The model mimics a functional periodontal ligament (PDL), which anchors dental roots to the jaw bone and transfers the axial load of mastication to the jaw bone. A collagen gel containing human PDL fibroblasts was seeded in a PDL space between an artificial root and bone surface. The effects of 3-day loading on the fibroblasts were studied in vitro by axial and intermittent displacement of the root to which the gel was attached. Cell responses were recorded by measuring expression of three sets of genes: (i) cyclooxygenase 1 and 2 (COX-1, COX-2) producing prostaglandins (signaling molecules); (ii) Runx2, a transcription factor for the osteogenic lineage; and (iii) the extracellular matrix proteins osteopontin, dentin matrix protein 1, and collagen type I (COL1). Loading for 3 days resulted in magnitude-dependent changes in the expression of COX-2 and COL1. A low loading magnitude significantly decreased COX-2 expression, an intermediate magnitude increased its expression, while a high magnitude increased COL1 expression. We concluded that the 3D loading model provides a useful, well-controlled method to examine ligament fibroblast responses to mechanical loading. The model may serve to explore the application of mechanical loading as an anabolic factor for ligament reconstruction.


Subject(s)
Models, Biological , Periodontal Ligament/physiology , Regeneration/physiology , Weight-Bearing/physiology , Adult , Benzophenones , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gels , Gene Expression Regulation/drug effects , Humans , Ketones/pharmacology , Male , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Periodontal Ligament/enzymology , Polyethylene Glycols/pharmacology , Polymers , Proline/metabolism , Regeneration/drug effects , Tissue Engineering , Young Adult
16.
Biomaterials ; 28(24): 3530-6, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17512583

ABSTRACT

Reconstruction of tendon and ligament tissues requires proper attachment of the tissue-engineered construct to surrounding tissues. A problem of reconstructing collagen-rich tissues is that an in vitro engineered collagenous network containing fibroblasts will contract and detach from a solid surface. In vivo anchorage of soft connective tissues to mineralized tissues like bones and teeth is accomplished by embedding collagen fibrils into mineralized layers. Mineralization is partially the result of local activity of the enzyme alkaline phosphatase (ALP). In this study, we tested whether ALP-induced mineral deposition at the interface between a collagen gel and a polystyrene or polyetheretherketone (PEEK) surface could prevent gel detachment from the surface. Coating of culture wells with intestinal ALP prevented detachment of gels harbored with human periodontal ligament (PDL) fibroblasts in the presence of its substrate beta-glycerophosphate. Mineral deposition was observed predominantly at the interface of collagen gel and well surface. The contractile properties of fibroblasts were not influenced by either ALP, beta-glycerophosphate or both. The presence of ALP on a solid surface and providing its substrate to allow mineral deposition can prevent detachment of collagen matrices. Our findings provide a tool to induce attachment of fibrillar collagen to a solid surface; an approach that seems useful for reconstruction of load-bearing tissues and attachment of ligaments to implants.


Subject(s)
Alkaline Phosphatase/metabolism , Collagen/metabolism , Adult , Calcium/metabolism , Cell Adhesion , Cells, Cultured , Fibroblasts/cytology , Humans , Male , Microscopy, Electron , Protein Binding , Tissue Engineering
17.
Matrix Biol ; 25(8): 515-22, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16973341

ABSTRACT

Extracellular matrix components play an important role in modulating cellular activity. To study such capacities of the matrix, fibroblasts are frequently cultured in a three-dimensional gel and contraction is assessed as a measure of cellular activity. Since a connective tissue contains several types of collagen, we investigated the effect of gels composed of collagen I alone or in combination with 10% collagen III and/or 5% collagen V on contraction by human periodontal ligament fibroblasts. Gels containing collagen V contracted much faster than those without this type of collagen. Blocking of the integrin beta1-subunit with an activity-blocking antibody delayed (gels with collagen V) or almost completely blocked (gels without collagen V) contraction. Use of an antibody directed against integrin alpha2beta1 resulted in delay of gel contraction for gels both with and without collagen V. Anti-integrin alpha v beta3 or RGD peptides partially blocked contraction of gels containing collagen V, but had no effect on gels consisting of collagen I alone. The beta1-containing integrins are involved in the basal contraction by fibroblasts that bind to collagens I and III. The enhanced contraction, stimulated by collagen V, appears to be mediated by integrin alpha v beta3. We conclude that collagen V may play an important modulating role in connective tissue contraction. Such a modulation may occur during the initial stages of wound healing and/or tissue regeneration.


Subject(s)
Collagen Type V/physiology , Collagen/metabolism , Fibroblasts/metabolism , Integrins/metabolism , Periodontal Ligament/metabolism , Adult , Collagen/chemistry , Collagen Type V/metabolism , Extracellular Matrix/metabolism , Humans , Integrin alpha2beta1/biosynthesis , Integrin alphaVbeta3/metabolism , Male , Regeneration , Time Factors , Wound Healing
18.
J Control Release ; 83(2): 241-51, 2002 Oct 04.
Article in English | MEDLINE | ID: mdl-12363450

ABSTRACT

Tumor vasculature can be targeted by peptides containing an RGD (Arg-Gly-Asp) sequence, which bind to alpha(v)beta3 and alpha(v)beta5 integrins on angiogenic endothelial cells. By covalently attaching cyclic RGD-peptides (cRGDfK) to a protein backbone, we prepared a multivalent peptide-protein conjugate with increased affinity for alpha(v)beta3/alpha(v)beta5 integrins. We demonstrated that RGDpep-protein conjugate bound to HUVEC, whereas the conjugate prepared with the control RAD peptide was devoid of any binding. RGDpep-protein conjugate was furthermore functional in inhibiting the adhesion of HUVEC to alpha(v)beta3/alpha(v)beta5 ligand vitronectin, and direct binding of the radiolabeled conjugate to HUVEC was inhibited by alpha(v)beta(3)/alpha(v)beta5-specific RGD peptides. Finally, RGDpep-protein conjugate was shown to be internalized and degraded by HUVEC, a process that could be inhibited by lysosomal degradation inhibitors chloroquine and ammonium chloride. This cellular handling was significantly influenced by the presence of cations, which strongly inhibited internalization. This is the first study that shows direct evidence that primary endothelial cells are capable of internalizing RGD-containing macromolecular proteins. This feature makes them attractive carriers for the intracellular delivery of potent anti-angiogenic drugs into endothelial cells for the treatment of cancer and chronic inflammatory diseases.


Subject(s)
Drug Delivery Systems/methods , Endothelium, Vascular/metabolism , Neoplasms/blood supply , Neoplasms/metabolism , Oligopeptides/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Oligopeptides/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...