Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Osteoarthritis Cartilage ; 24(5): 892-901, 2016 May.
Article in English | MEDLINE | ID: mdl-26687824

ABSTRACT

OBJECTIVE: To determine the strain-induced signaling pathways involved in regulating the transactivation of the transcription regulator Cbp/p300 Interacting Transactivator with ED-rich tail 2 (CITED2) and downstream targets in chondrocytes. METHODS: Primary human chondrocytes or C28/I2 chondrocytic cells were subjected to various strain regimes. C57BL/6 mice were subjected to treadmill running. Loss-of-function was carried out using siRNA or inhibitors specific for targeted molecules. mRNA levels were assayed by RT-qPCR, and proteins by western blotting, immunofluorescence, and/or immunohistochemical staining. CITED2 promoter activity was assayed in chondrocytes using wild-type or mutant constructs. RESULTS: Cyclic strain at 5%, 1 Hz induced CITED2 expression and suppressed expression of matrix metalloproteinase (MMP)-1 and -13 at the messenger RNA (mRNA) and protein levels in human chondrocytes. Abolishing primary cilia through knockdown of intraflagellar transport protein (IFT88) attenuated CITED2 gene expression and decreased protein levels. Similar effects were observed with inhibitors of extracellular adenosine triphosphate (ATP) or P2 purinergic receptors, or antagonists of Ca(2+) signaling. Knockdown of IFT88 in articular chondrocytes in vivo diminished treadmill induced-CITED2 expression and upregulated MMPs. Knockdown of hypoxia-inducible factor (HIF)1α, specificity protein 1 (Sp1), or deletion of the shear stress response element (SSRE) in the CITED2 promoter limited cyclic strain-induced transactivation of CITED2. However, the strain induced-transactivation of CITED2 was abolished only on knockdown of HIF1α, Sp1, and SSRE or by loss-of-function of IFT88 or extracellular-signal-regulated kinases (ERK)1/2. CONCLUSIONS: CITED2 transactivation is a critical event in signaling generated by strain and transduced by primary cilia, extracellular ATP, P2 purinergic receptors, and Ca(2+) signaling. Strain-induced CITED2 transactivation requires HIF1α, Sp1, and an intact SSRE and leads to the downregulation of MMPs such as MMP-1 and MMP-13.


Subject(s)
Adenosine Triphosphate/physiology , Calcium Signaling/physiology , Chondrocytes/metabolism , Mechanotransduction, Cellular/physiology , Repressor Proteins/biosynthesis , Trans-Activators/biosynthesis , Animals , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes/physiology , Cilia/metabolism , Down-Regulation , Humans , MAP Kinase Signaling System/physiology , Male , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 13/genetics , Mice, Inbred C57BL , Physical Exertion/physiology , RNA, Messenger/genetics , Repressor Proteins/genetics , Trans-Activators/genetics , Transcriptional Activation/physiology
2.
J Mech Behav Biomed Mater ; 28: 183-94, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23994943

ABSTRACT

Osteocytes are terminally differentiated bone cells, derived from osteoblasts, which are vital for the regulation of bone formation and resorption. ECM stiffness and cell seeding density have been shown to regulate osteoblast differentiation, but the precise cues that initiate osteoblast-osteocyte differentiation are not yet understood. In this study, we cultured MC3T3-E1 cells on (A) substrates of different chemical compositions and stiffnesses, as well as, (B) substrates of identical chemical composition but different stiffnesses. The effect of cell separation was investigated by seeding cells at different densities on each substrate. Cells were evaluated for morphology, alkaline phosphatase (ALP), matrix mineralisation, osteoblast specific genes (Type 1 collagen, Osteoblast specific factor (OSF-2)), and osteocyte specific proteins (dentin matrix protein 1 (DMP-1), sclerostin (Sost)). We found that osteocyte differentiation (confirmed by dendritic morphology, mineralisation, reduced ALP, Col type 1 and OSF-2 and increased DMP-1 and Sost expression) was significantly increased on soft collagen based substrates, at low seeding densities compared to cells on stiffer substrates or those plated at high seeding density. We propose that the physical nature of the ECM and the necessity for cells to establish a communication network contribute substantially to a concerted shift toward an osteocyte-like phenotype by osteoblasts in vitro.


Subject(s)
Cell Differentiation , Extracellular Matrix/metabolism , Mechanical Phenomena , Osteocytes/cytology , 3T3 Cells , Alkaline Phosphatase/metabolism , Animals , Biomechanical Phenomena , Gene Expression Regulation , Mice , Minerals/metabolism , Osteocytes/metabolism
3.
Osteoarthritis Cartilage ; 20(9): 1011-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22613702

ABSTRACT

OBJECTIVE: Recent developments on high resolution micro computed tomography (µCT) allow imaging of soft tissues in small animal joints. Nevertheless, µCT images cannot distinguish soft tissues from synovial fluid due to their similar mass density, limiting the 3D assessment of soft tissues volume and thickness. This study aimed to evaluate a lead chromate contrast agent for µCΤ arthrography of rat knee joints ex vivo. DESIGN: Intact tibiofemoral rat joints were injected with the contrast agent at different concentrations and imaged using a µCT at 2.7 µm isotropic voxel size. Cartilage thickness was measured using an automated procedure, validated against histological measurements, and analyzed as a function of µCT image resolution. Changes in hard and soft tissues were also analyzed in tibiofemoral joints 4 weeks after surgical destabilization of the medial meniscus (DMM). RESULTS: The contrast agent diffused well throughout the whole knee cavity without penetrating the tissues, therefore providing high contrast at the boundaries between soft tissues and synovial fluid space. Thickness analysis of cartilage demonstrated a high similarity between histology and µ-arthrography approaches (R(2) = 0.90). Four weeks after surgical DMM, the development of osteophytes (Oph) and cartilage ulcerations was recognizable with µCT, as well as a slight increase in trabecular bone porosity, and decrease in trabecular thickness. CONCLUSIONS: A lead chromate-based contrast agent allowed discriminating the synovial fluid from soft tissues of intact knee joints, and thus made possible both qualitative and quantitative assessment of hard and soft tissues in both intact and DMM tibiofemoral joints using high resolution µCT.


Subject(s)
Arthrography/methods , Cartilage, Articular/diagnostic imaging , Chromates , Contrast Media , Hindlimb/diagnostic imaging , Lead , Animals , Cartilage, Articular/pathology , Female , Hindlimb/pathology , Menisci, Tibial/diagnostic imaging , Osteophyte/diagnostic imaging , Rats , Rats, Sprague-Dawley
4.
Bone ; 47(4): 766-72, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20633708

ABSTRACT

Recent experiments point to two predominant forms of fatigue microdamage in bone: linear microcracks (tens to a few hundred microns in length) and "diffuse damage" (patches of diffuse stain uptake in fatigued bone comprised of clusters of sublamellar-sized cracks). The physiological relevance of diffuse damage in activating bone remodeling is not known. In this study microdamage amount and type were varied to assess whether linear or diffuse microdamage has similar effects on the activation of intracortical resorption. Activation of resorption was correlated to the number of linear microcracks (Cr.Dn) in the bone (R(2)=0.60, p<0.01). In contrast, there was no activation of resorption in response to diffuse microdamage alone. Furthermore, there was no significant change in osteocyte viability in response to diffuse microdamage, suggesting that osteocyte apoptosis, which is known to activate remodeling at typical linear microcracks in bone, does not result from sublamellar damage. These findings indicate that inability of diffuse microdamage to activate resorption may be due to lack of a focal injury response. Finally, we found that duration of loading does not affect the remodeling response. In conclusion, our data indicate that osteocytes activate resorption in response to linear microcracks but not diffuse microdamage, perhaps due to lack of a focal injury-induced apoptotic response.


Subject(s)
Bone Remodeling/physiology , Fatigue/physiopathology , Stress, Mechanical , Ulna/pathology , Ulna/physiopathology , Animals , Fatigue/pathology , Osteocytes/pathology , Rats , Rats, Sprague-Dawley
5.
Bone ; 46(3): 577-83, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19925896

ABSTRACT

INTRODUCTION: Osteocyte apoptosis has been linked to bone resorption resulting from estrogen depletion and other resorptive stimuli; however, precise spatial and temporal relationships between the two events have not been clearly established. The purpose of this study was to characterize the patterns of osteocyte apoptosis in relation to bone resorption following ovariectomy to test whether osteocyte apoptosis occurs preferentially in areas known to activate resorption. Moreover, we report that osteocyte apoptosis is necessary to initiate endocortical remodeling in response to estrogen withdrawal. MATERIALS AND METHODS: Adult female C57BL/6J mice (17 weeks old) underwent either bilateral ovariectomy (OVX), or sham surgery (SHAM) and were euthanized on days 3, 7, 14, or 21 days after OVX. Diaphyseal cross-sections were stained by immunohistochemistry for activated caspase-3 as a marker of apoptosis. The percentages of caspase-positive stained osteocytes (Casp+Ot.) were measured along major and minor anatomical axes around the femoral diaphysis to evaluate the distribution of osteocyte apoptosis after estrogen loss; resorption surface was measured at the adjacent endocortical regions. In a second study to test whether osteocyte apoptosis plays a regulatory role in the initiation of bone resorption, a group of OVX mice received the pan-caspase inhibitor, QVDOPh, to inhibit osteocyte apoptosis. Remaining experimental and sham groups received either QVD or Vehicle. RESULTS: OVX increased osteocyte apoptosis in a non-uniform distribution throughout the femoral diaphyses. Increases in Casp+osteocytes were predominantly located in the posterior diaphyseal cortex. Here, the number of apoptotic osteocytes 4- to 7-fold higher than sham controls (p<0.005) by day 3 post-OVX and remained elevated. Increases in resorption post-OVX also occurred along the posterior endocortical surface overlying the region of osteocyte apoptosis, but these increases occurred only at 14 and 21 days post-OVX (p<0.002) well after the increases in osteocyte apoptosis. Treatment with QVD in OVX animals suppressed osteocyte apoptosis, with levels in QVD-treated samples equivalent to baseline. Moreover, the increases in osteoclastic resorption normally observed after estrogen loss did not occur in OVX mice treated with QVD. CONCLUSIONS: The results of this study demonstrate that osteocyte apoptosis following estrogen loss occur regionally, rather than uniformly throughout the cortex. We also showed that estrogen loss increased osteocyte apoptosis. Apoptotic osteocytes were overwhelmingly localized within the posterior cortical region, the location where endocortical resorption was subsequently activated in ovariectomized mice. Finally, the increases in osteoclastic resorption normally observed after estrogen withdrawal did not occur in the absence of osteocyte apoptosis indicating that this apoptosis is necessary to activate endocortical remodeling following estrogen loss.


Subject(s)
Apoptosis/physiology , Bone Resorption/pathology , Osteocytes/pathology , Ovariectomy , Animals , Bone Resorption/metabolism , Bone Resorption/physiopathology , Female , Mice , Mice, Inbred C57BL , Osteocytes/metabolism , Osteocytes/physiology , Ovariectomy/adverse effects , Time Factors
6.
Anat Rec (Hoboken) ; 292(3): 355-63, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19248169

ABSTRACT

In order for osteocytes to perceive mechanical information and regulate bone remodeling accordingly they must be anchored to their extracellular matrix (ECM). To date the nature of this attachment is not understood. Osteocytes are embedded in mineralized bone matrix, but maintain a pericellular space (50-80 nm) to facilitate fluid flow and transport of metabolites. This provides a spatial limit for their attachment to bone matrix. Integrins are cell adhesion proteins that may play a role in osteocyte attachment. However, integrin attachments require proximity between the ECM, cell membrane, and cytoskeleton, which conflicts with the osteocytes requirement for a pericellular fluid space. In this study, we hypothesize that the challenge for osteocytes to attach to surrounding bone matrix, while also maintaining fluid-filled pericellular space, requires different "engineering" solutions than in other tissues that are not similarly constrained. Using novel rapid fixation techniques, to improve cell membrane and matrix protein preservation, and transmission electron microscopy, the attachment of osteocyte processes to their canalicular boundaries are quantified. We report that the canalicular wall is wave-like with periodic conical protrusions extending into the pericellular space. By immunohistochemistry we identify that the integrin alphavbeta3 may play a role in attachment at these complexes; a punctate pattern of staining of beta3 along the canalicular wall was consistent with observations of periodic protrusions extending into the pericellular space. We propose that during osteocyte attachment the pericellular space is periodically interrupted by underlying collagen fibrils that attach directly to the cell process membrane via integrin-attachments.


Subject(s)
Bone Matrix/physiology , Cell Membrane/ultrastructure , Integrin alphaVbeta3/metabolism , Osteocytes/physiology , Animals , Bone Matrix/ultrastructure , Cell Adhesion/physiology , Cell Membrane/metabolism , Cell Physiological Phenomena/physiology , Fluorescence , Immunoenzyme Techniques , Mice , Osteocytes/ultrastructure
7.
Bone ; 35(5): 1095-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15542034

ABSTRACT

Woven bone forms rapidly during tissue growth, following injury and in response to certain anabolic stimuli. Functional differences between woven and lamellar bone may be due, in part, to differences in osteocyte density (cells per unit tissue). Woven bone has been estimated to contain four to eight times more osteocytes than lamellar bone, although primary data to support this assertion are limited. Given recent findings implicating osteocytes as regulators of bone remodeling, bone formation and bone volume, such large differences in osteocyte density between woven and lamellar bone may have important consequences. In this study, we compared the density of osteocyte lacunae (lacunae/mm(2) tissue) in rat lamellar bone with that in woven bone formed under several different circumstances. We found that the lacunar density of lamellar cortical bone in the rat (834+/-83 cells/mm2, mean+/-SD) did not differ significantly from that of periosteal woven bone formed via intramembranous osteogenesis, either in response to mechanical loading (921+/-204 cells/mm2) or in the periosteal buttressing region of the fracture callus (1138+/-168 cells/mm2). In contrast, lacunar density of endochondrally derived woven bone in the center (gap) region of fracture callus was nearly 100% greater (1875+/-270 cells/mm2) than in lamellar cortical bone while lacunar density of primary spongiosa of the growth plate was 40% greater (1674+/-228 cells/mm2) than that in lamellar cancellous bone (1189+/-164). These findings demonstrate that lacunar density in woven bone varies depending on skeletal site and developmental history and appears to be elevated in endochondrally derived woven bone adjacent to marrow space. Given the considerable evidence supporting osteocytes as local initiators of bone remodeling, we suggest that woven bone with increased lacunar density may undergo remodeling at an accelerated rate.


Subject(s)
Bone Remodeling/physiology , Bone and Bones/cytology , Osteocytes/cytology , Animals , Bone Development/physiology , Bone Regeneration/physiology , Bony Callus/cytology , Cell Count , Female , Femoral Fractures/pathology , Femur/cytology , Male , Microscopy, Confocal , Rats , Rats, Sprague-Dawley , Tibia/cytology , Ulna/cytology , Weight-Bearing/physiology
8.
Bone ; 29(6): 560-4, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11728927

ABSTRACT

Angiogenesis is considered essential to fracture healing, but its role in the healing process remains poorly understood. Angiogenesis inhibitors, which block new blood vessel formation by specifically targeting vascular cells, are currently under development for use in cancer chemotherapy, and are potentially powerful tools for defining the consequences of angiogenic impairment on fracture healing. In this study, we directly tested the effects of the angiogenesis inhibitor TNP-470 on the healing of closed femoral fractures in an established rat model system. Beginning 1 day after fracture, animals received either angiogenesis inhibitor at a therapeutically effective antitumor dose, or a weight-adjusted amount of carrier vehicle. The progress of fracture healing was assessed at weekly intervals for 21 days by radiography and histology; functional assessment was carried out at day 24 by biomechanical testing. By all three criteria, treatment with the angiogenesis inhibitor completely prevented fracture healing. Formation of both callus and periosteal woven bone were suppressed, indicating that both the intramembranous and endochondral pathways of osteogenesis were affected. The resulting tissue resembled "atrophic nonunions" often seen clinically in cases of failed fracture healing, but rarely achieved in animal models. These results show that angiogenesis is essential to very early stages of fracture healing, and suggest this model system may be useful for understanding the mechanisms underlying fracture nonunions due to vascular impairment. Finally, the data raise the possibility that impairment of fracture healing may be an adverse effect of clinical treatments with antiangiogenic drugs.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Fracture Healing/drug effects , Sesquiterpenes/pharmacology , Animals , Cyclohexanes , Female , Femur/pathology , O-(Chloroacetylcarbamoyl)fumagillol , Rats , Rats, Sprague-Dawley
9.
J Orthop Res ; 19(3): 339-45, 2001 May.
Article in English | MEDLINE | ID: mdl-11398843

ABSTRACT

The proto-oncogene c-fos has been implicated in the development of both benign and malignant lesions of bone. Although c-fos expression in such lesions has been well studied in transgenic mouse models, less is known about its role in human musculoskeletal pathology. To clarify this relationship, we used in situ hybridization to localize c-fos m-RNA transcripts in 26 fibrous lesions (eight cases of extra-abdominal fibromatosis and six cases each of fibrous dysplasia, fibrosarcoma, and malignant fibrous histiocytoma of bone) as well as six chondrosarcomas and eight conventional high grade osteosarcomas. We found detectable levels of c-fos expression in tissues from each type of lesion tested. Moreover, all fibrous lesions consistently demonstrated high levels of expression in a majority of cells in each lesion. Chondrosarcomas and osteosarcomas exhibited more heterogeneity in c-fos expression than fibrous tissues. Three of six chondrosarcomas showed moderate expression of c-fos while only one of six was considered high. Similarly, only three of eight osteosarcomas had high expression of c-fos. These findings indicate that the expression of c-fos may be important in the development of a broad range of fibrous lesions as well as in bone and cartilaginous tumors. Additionally, this is the first report, to our knowledge, of detectable c-fos m-RNA in human chondrosarcoma.


Subject(s)
Bone Neoplasms/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Child , Child, Preschool , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , Female , Fibroma/genetics , Fibroma/metabolism , Fibroma/pathology , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Fibrous Dysplasia of Bone/genetics , Fibrous Dysplasia of Bone/metabolism , Fibrous Dysplasia of Bone/pathology , Histiocytoma, Benign Fibrous/genetics , Histiocytoma, Benign Fibrous/metabolism , Histiocytoma, Benign Fibrous/pathology , Humans , In Situ Hybridization , Male , Middle Aged , Neoplasm Staging , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/metabolism
10.
Endocrinology ; 140(9): 3904-8, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10465258

ABSTRACT

Maintenance of uterine-placental attachment during human pregnancy may depend at least partly on adhesive interactions between cytotrophoblasts and their extracellular matrix (ECM). Such interactions are often mediated by integrins, signal-transducing heterodimeric transmembrane glycoproteins. We previously showed that glucocorticoid (GC) suppressed the expression of collagen and laminin in human placenta; here we show that GC also modulates the expression by human cytotrophoblasts of the integrin subunits alpha2 and beta1, components of a known receptor for these ECM ligands. Cytotrophoblasts were isolated from human term placentas, cultured up to 4 days in the presence of 0-1000 nM dexamethasone (DEX), and assayed for 1) integrin messenger RNA (mRNA) levels by Northern hybridization, 2) integrin subunit synthesis after [35S]methionine labeling, or 3) cell surface integrin levels after 125I labeling by lactoperoxidase. In four independent experiments, 100 nM DEX reduced mRNA levels for integrin alpha2 to 6+/-1% of the control value. This effect was similar between 1-4 days of treatment and was dose dependent between 1-1000 nM DEX. Cortisol treatment (100 nM) inhibited levels of integrin alpha2 mRNA, but 100 nM testosterone, estradiol, and progesterone were less effective, suggesting that this response was specific to GC. In immunoprecipitation studies, treatment of cytotrophoblasts with 100 nM DEX for 2 days reduced the rates of synthesis of the alpha2 integrin subunit as well as its expression on the cell surface to 1-10% of control levels. DEX effects on the beta1 integrin subunit were less dramatic. DEX reduced beta1 mRNA levels to only 69+/-8% of control levels, a smaller reduction compared with effects on alpha2 integrin mRNA. DEX inhibited beta1 protein synthesis and cell surface expression to 60-70% of control levels. In all experiments, DEX had no effect on total protein synthesis. Thus, our results demonstrate that GC treatment specifically and markedly down-regulates expression of alpha2 integrin subunit by human cytotrophoblasts. This finding is consistent with the concept that uterine-placental adherence across gestation may be regulated by coordinate effects on ECM ligands and cellular adhesion receptors.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Integrins/metabolism , Placenta/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cells, Cultured , Female , Humans , Integrin alpha2 , Pregnancy , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Trophoblasts/drug effects , Trophoblasts/metabolism
11.
J Bone Joint Surg Br ; 81(3): 508-15, 1999 May.
Article in English | MEDLINE | ID: mdl-10872376

ABSTRACT

The multifunctional adhesion molecule CD44 is a major cell-surface receptor for hyaluronic acid (HUA). Recent data suggest that it may also bind the ubiquitous bone-matrix protein, osteopontin (OPN). Because OPN has been shown to be a potentially important protein in bone remodelling, we investigated the hypothesis that OPN interactions with the CD44 receptor on bone cells participate in the regulation of the healing of fractures. We examined the spatial and temporal patterns of expression of OPN and CD44 in healing fractures of rat femora by in situ hybridisation and immunohistochemistry. We also localised HUA in the fracture callus using biotinylated HUA-binding protein. OPN was expressed in remodelling areas of the hard callus and was found in osteocytes, osteoclasts and osteoprogenitor cells, but not in cuboidal osteoblasts which were otherwise shown to express osteocalcin. The OPN signal in osteocytes was not uniformly distributed, but was restricted to specific regions near sites where OPN mRNA-positive osteoclasts were attached to bone surfaces. In the remodelling callus, intense immunostaining for CD44 was detected in osteocyte lacunae, along canaliculi, and on the basolateral plasma membrane of osteoclasts, but not in the cuboidal osteoblasts. HUA staining was detected in fibrous tissues but little was observed in areas of hard callus where bone remodelling was progressing. Our findings suggest that OPN, rather than HUA, is the major ligand for CD44 on bone cells in the remodelling phase of healing of fractures. They also raise the possibility that such interactions may be involved in the communication of osteocytes with each other and with osteoclasts on bone surfaces. The interactions between CD44 and OPN may have important clinical implications in the repair of skeletal tissues.


Subject(s)
Bony Callus/pathology , Femoral Fractures/pathology , Fracture Healing/physiology , Hyaluronan Receptors/analysis , Sialoglycoproteins/analysis , Animals , Bone Remodeling/physiology , Hyaluronic Acid/analysis , Male , Osteoclasts/pathology , Osteocytes/pathology , Osteopontin , Rats , Rats, Sprague-Dawley
12.
Clin Orthop Relat Res ; (366): 258-63, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10627743

ABSTRACT

This investigation tested the hypothesis that daily parenterally administered parathyroid hormone (1-34) improves fracture healing. Twenty, 3-month-old, male Sprague Dawley rats weighing approximately 400 g each, underwent the production of closed, unilateral mid-diaphyseal femoral fractures. Animals were divided into two groups of 10; the animals received either a daily subcutaneous injection of delivery vehicle (0.9% saline) or 80 micrograms/kg parathyroid hormone. On Day 21 after fracture the animals were euthanized, the femurs were removed and subjected to biomechanical testing, bone densitometry (dual energy x-ray absorptiometry, peripheral quantitative computed tomography), and histologic examination. Treatment with parathyroid hormone resulted in statistically significant increases in callus area and strength. Histologic examination of the calluses showed an increase in the amount of new bone formed. No differences were observed in the weights of the animals or the sizes of the bones. Values obtained using dual energy x-ray absorptiometry and peripheral quantitative computed tomography indicate an increase in density in the parathyroid hormone treated fractures consistent with the histologic appearance and the findings of increased strength, although these bone density changes did not achieve statistical significance. These results suggest that parenterally administered parathyroid hormone (1-34) may enhance or accelerate normal fracture healing and support the concept that this hormone be tested clinically as a systemic treatment for fractures that are slow to heal.


Subject(s)
Femoral Fractures/physiopathology , Fracture Healing/drug effects , Fractures, Closed/physiopathology , Parathyroid Hormone/therapeutic use , Absorptiometry, Photon , Animals , Body Weight , Bone Density , Bony Callus/drug effects , Bony Callus/pathology , Bony Callus/physiopathology , Cartilage/drug effects , Cartilage/pathology , Cartilage/physiopathology , Elasticity , Femoral Fractures/diagnostic imaging , Femoral Fractures/pathology , Fractures, Closed/diagnostic imaging , Fractures, Closed/pathology , Injections, Subcutaneous , Male , Osteogenesis/drug effects , Osteogenesis/physiology , Parathyroid Hormone/administration & dosage , Pharmaceutical Vehicles , Rats , Rats, Sprague-Dawley , Stress, Mechanical , Tomography, X-Ray Computed , Torsion Abnormality
13.
J Orthop Trauma ; 12(2): 111-6, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9503300

ABSTRACT

OBJECTIVES: (a) To develop a reliable and reproducible system for distraction osteogenesis in the rat to establish a model for future investigations of bone repair and regeneration. (b) To describe and characterize the histological events in distraction osteogenesis in the rat and to determine whether cartilage development is a normal component of the process. STUDY DESIGN: Species-specific, longitudinal time study. METHODS: Twenty rats underwent production of a middiaphyseal femoral osteotomy and application of a monolateral external fixator specifically designed for distraction. Animals were divided into five groups based on the time and extent of lengthening. RESULTS: During distraction, gap tissue showed collagen bundles and fibroblasts that were oriented longitudinally to the direction of the distraction force. Woven bone appeared to be laid down on these collagen scaffolds, and the newly formed vascular sinuses appeared to be the sites from which bone formation was initiated within the distraction gap. All groups undergoing active distraction showed intramembranous ossification in the distraction gap and endochondral ossification peripherally. However, when distraction was discontinued, endochondral ossification was observed in the gap. CONCLUSION: Distraction produces an environment in the distraction gap that suppresses the formation of cartilage. The formation of cartilage by injured periosteum, however, is obligatory and does not appear to be influenced by distraction. Bone formation within the distraction gap occurs where angiogenesis develops.


Subject(s)
Bone Regeneration , Cartilage/physiology , Disease Models, Animal , Osteogenesis, Distraction , Osteogenesis , Animals , Femur/surgery , Male , Osteotomy , Rats , Rats, Sprague-Dawley , Reproducibility of Results
14.
Clin Orthop Relat Res ; (340): 267-75, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9224265

ABSTRACT

Direct physical injury to bone marrow is associated with a systemic osteogenic response. However, blood loss, a condition that stimulates hemopoietic stem cells, also may activate osteoprogenitor cells in the bone marrow. To determine if bleeding induces a systemic osteogenic response, the mineral appositional rates and osteoblast numbers were determined in the bones of rats that were subjected to controlled cardiac bleeding and compared with those of rats subjected to ablation of their tibial bone marrow. In addition, a study of the kinetics of the osteogenic responses during the first 10 days after operative treatment was performed by quantitating the serum levels of biochemical indices known to be associated with systemic bone formation. The results showed that animals that sustained acute blood loss (1% or 3% body weight) or injury to their tibial bone marrow had statistically significant increases in mineral appositional rate, osteoblast number, and serum levels of osteogenic growth peptide. The kinetics studies showed that osteogenic growth peptide levels peaked on the tenth postoperative day and declined sharply thereafter. An enhancement of serum osteocalcin activity occurred only on the second postoperative day, was increased in all experimental groups when compared with untreated control animals, but immediately declined to baseline levels. Alkaline phosphatase activities increased in the experimental groups, peaking on Day 10 after tibial bone marrow ablation and on Day 12 in the group that underwent bleeding. These findings suggest that bleeding alone, independent of any skeletal trauma, may evoke a systemic osteogenic response. This response is similar in its timing and magnitude to that which has been shown to follow direct physical injury to bone marrow. The observation that systemic bone formation follows bone marrow activation induced by two different stimuli suggests that these responses may be mediated by common regulatory mechanisms. The ability to trigger or control these responses may form the basis for future therapeutic strategies to enhance bone formation.


Subject(s)
Bone Remodeling/physiology , Hematopoiesis/physiology , Hemorrhage/physiopathology , Intercellular Signaling Peptides and Proteins , Alkaline Phosphatase/blood , Animals , Enzyme-Linked Immunosorbent Assay , Growth Substances/blood , Histones , Male , Osteocalcin/blood , Osteogenesis/physiology , Peptides/blood , Phlebotomy , Rats , Rats, Sprague-Dawley
15.
J Orthop Res ; 15(5): 757-64, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9420607

ABSTRACT

Skeletal development involves the coordinated participation of several types of collagen, including both major and minor fibrillar collagens. Although much is known about the major fibrillar collagens, such as types I and II, less is known about the minor fibrillar collagens, and their role in the repair and regeneration of bone has not been extensively studied. To clarify the role of minor fibrillar collagens in fracture repair, we examined the spatial and temporal expression of mRNAs for pro-alpha 2(V) collagen and pro-alpha 1(XI) collagen in healing fractures in the rat by in situ hybridization and compared their patterns of expression with those of mRNAs for pro-alpha 1(I) collagen, pro-alpha 1(II) collagen, and osteocalcin. A strong signal for pro-alpha 2(V) was detected in the periosteal osteoprogenitor cells, whereas osteocalcin mRNA was strongly expressed only in the deep layers of the hard callus. The distribution of the pro-alpha 2(V) signal was correlated with that of pro-alpha 1(I) but was mutually exclusive of that of pro-alpha 1(II). The expression of pro-alpha 1(XI) mRNA was synchronously regulated with that of pro-alpha 1(II) during chondrogenesis in the soft callus. In the hard callus, pro-alpha 1(XI) signal was found in osteoblastic cells at the site of intramembranous and endochondral ossification. These cells simultaneously expressed pro-alpha 2(V), although they were negative for pro-alpha 1(II). These findings suggest that the alpha 2(V) collagen chain participates in the formation of the noncartilaginous fibrillar network in the hard callus and preferentially contributes to the initial stage of the intramembranous bone formation. Recent reports have revealed that type-XI collagen, which had been classified as a cartilage-type collagen, is not necessarily specific for cartilage. The present results advanced this recognition and demonstrated a coexpression of alpha 1(XI) mRNA and alpha 2(V) mRNA in the noncartilaginous tissues in the fracture callus; this suggests the presence of tissue-specific and stage-specific heterotrimers consisting of alpha 1(XI) and alpha 2(V) collagen chains and the association of such hybrid trimers with the major fibrillar collagens in the process of fracture healing.


Subject(s)
Collagen/metabolism , Femoral Fractures/metabolism , Femur/injuries , Fracture Healing , Gene Expression Regulation , Procollagen/metabolism , Animals , Bony Callus/diagnostic imaging , Bony Callus/metabolism , Bony Callus/pathology , Chondrocytes/metabolism , Collagen/genetics , Femoral Fractures/diagnostic imaging , Femoral Fractures/pathology , Femur/diagnostic imaging , Femur/metabolism , Femur/pathology , In Situ Hybridization , Male , Osteocalcin/genetics , Osteocalcin/metabolism , Procollagen/genetics , RNA, Messenger/metabolism , Radiography , Rats , Rats, Sprague-Dawley
16.
J Bone Miner Res ; 10(8): 1272-81, 1995 Aug.
Article in English | MEDLINE | ID: mdl-8585432

ABSTRACT

Cytokines, a group of proteins known to regulate hemopoietic and immune functions, are also involved in inflammation, angiogenesis, and bone and cartilage metabolism. Since all of these processes occur following bone injury, or are known to contribute to wound repair mechanisms, this investigation sought to test the hypothesis that cytokines are involved in fracture healing. Two sets of 60 male Sprague-Dawley rats underwent the production of standard closed femoral fractures. The animals were then euthanized in groups of 15 on days 3, 7, 14, and 21 postfracture. A separate control group was also used for the harvesting of intact unfractured bone. At the time of euthanasia, calluses or bone specimens were explanted to organ culture and treated with either media alone or media containing the inducing agents lipopolysaccharide or concanavalin A. A titration of conditioned medium from these cultures was then added to factor-dependent clonal cell lines that are known to be specifically responsive to interleukin-1, interleukin-6, granulocyte-macrophage colony stimulating factor or macrophage-colony stimulating factor. To confirm the identities of each of these cytokines, neutralizing antibody studies were performed. The results showed that interleukin-1 is expressed at very low constitutive levels throughout the period of fracture healing but can be induced to high activities in the early inflammatory phase (day 3). Granulocyte-macrophage colony stimulating factor showed no constitutive activity but could also be induced to high activities with lipopolysaccharide. The ability of these two cytokines to be induced declined progressively as fracture healing proceeded. Interleukin-6 showed high constitutive activity early in the healing process (day 3), and treatment with inducing agent did not increase the activity of this cytokine at this timepoint. Lipopolysaccharide did increase interleukin-6 activity in day 7 and 14 fracture calluses. Although macrophage-colony stimulating factor is thought to be involved in a variety of metabolic bone conditions, it could not be detected or induced from any of the callus samples. Moreover, none of the samples of unfractured bone showed constitutive or inducible activities for any of these cytokines. A separate experiment in which calluses and samples of unfractured bone from similar cultures were examined histologically and tested for DNA or protein synthesis at two timepoints in the culture period (days 1 and 4) showed that tissue viability was maintained. Thus the inability to detect macrophage colony-stimulating factor in fracture callus or any cytokine activity in unfractured bones was not due to cell death.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Bony Callus/metabolism , Cytokines/biosynthesis , Fracture Healing/physiology , Animals , Cell Line , Concanavalin A , DNA/biosynthesis , Disease Models, Animal , Femoral Fractures/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Interleukin-1/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides , Macrophage Colony-Stimulating Factor/biosynthesis , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
17.
Curr Opin Orthop ; 6(5): 7-13, 1995.
Article in English | MEDLINE | ID: mdl-11540472

ABSTRACT

Fracture healing is largely controlled by local regulatory interactions among cells and tissues near the site of injury; however, many systemic hormones including insulin, the glucocorticoids, and the gonadal steroids also can influence the course of tissue repair, particularly in the case of pathologic hormone excess or deficiency. Using well-defined animal models, recent studies have established that deficiencies in insulin and estrogen impair fracture healing, but data from this type of experiment are limited. Still, the similarities between morphogenetic events in fracture healing and those found in normal bone development and remodeling suggest that testable predictions can be made concerning hormonal effects on the progress of fracture healing. One concept that has received some direct experimental support in fracture healing model studies is that systemic hormones exert pleiotropic effects on callus tissue by regulating the expression and activity of local growth factors. Further verification of this and other predicted hormone effects should increase our understanding of the fundamental mechanisms underlying fracture repair, and may aid development of means to improve fracture healing in states of altered endocrine function.


Subject(s)
Androgens/metabolism , Diabetes Mellitus/metabolism , Estrogens/metabolism , Fracture Healing/physiology , Glucocorticoids/metabolism , Insulin/metabolism , Animals , Collagen/biosynthesis , Diabetes Mellitus/physiopathology , Disease Models, Animal , Fibroblast Growth Factor 2/metabolism , Fracture Healing/drug effects , Insulin/pharmacology , Rats , Somatomedins/metabolism
18.
J Bone Miner Res ; 9(9): 1441-9, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7817829

ABSTRACT

Synthetic Arg-Gly-Asp (RGD)-containing peptides were examined in bone resorption or attachment and detachment assays with isolated mammalian osteoclasts in an effort to elucidate the mechanistic and structural basis for the inhibition of bone resorption by s-echistatin. Bone resorption was the process most sensitive to inhibition by s-echistatin, with IC50 = 0.3 nM; inhibition of attachment to bone or detachment (lamellipodial retraction) was 30- to 70-fold less sensitive, with IC50 = 10 or 20 nM, respectively. Single amino acid substitutions within the 49-residue sequence of s-echistatin showed that although the efficacy of s-echistatin is dependent on the Arg24-Gly25-Asp26 sequence, additional residues, including Asp27, Met28, and Cys39, are also critical for potent inhibition of the resorbing activity of isolated rat osteoclasts. Because of the identification of the av beta 3 as the primary integrin on rat osteoclasts interacting the RGD peptides (Helfrich et al.), we examined the possibility of modeling bone resorption with other beta 3-mediated processes. Specifically, av beta 3 endothelial cell (human or rat) attachment to vitronectin and aIIb beta 3 platelet aggregation were compared with bone resorption for sensitivity to s-echistatin analogs, linear RGD peptides, and cyclic RGD peptides. Essentially no similarity in sensitivity to RGD peptides were observed between bone resorption, platelet aggregation, or endothelial cell attachment. Because rat osteoclasts and human giant cell tumors (osteoclastomas) shared similar sensitivity to s-echistatin and rat and human endothelial cells showed a similar sensitivity profile to RGD peptides, the dissimilarity of bone resorption to other beta 3-mediated processes cannot be explained in terms of species differences.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Bone Resorption/drug therapy , Oligopeptides/chemistry , Osteoclasts/drug effects , Peptides , Platelet Aggregation Inhibitors/therapeutic use , Viper Venoms/therapeutic use , Amino Acid Sequence , Animals , Bone Neoplasms/pathology , Cell Adhesion/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Giant Cell Tumor of Bone/pathology , Humans , Intercellular Signaling Peptides and Proteins , Mice , Microscopy, Confocal , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Molecular Sequence Data , Organ Culture Techniques , Osteoclasts/cytology , Osteoclasts/ultrastructure , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Viper Venoms/chemistry , Viper Venoms/pharmacology
19.
J Bone Joint Surg Am ; 76(5): 713-21, 1994 May.
Article in English | MEDLINE | ID: mdl-8175820

ABSTRACT

Estrogens play an important but poorly understood role in the maintenance of skeletal mass. Whereas the mechanisms of estrogen action on bone may be complex, the finding that osteoblasts express estrogen receptors suggests that this class of hormones exerts direct effects on bone cells. To understand how estrogens regulate osteoblastic function, the physiologically active estrogen metabolite 17 beta-estradiol was tested to determine its effects on the well characterized murine osteoblastic cell-line MC3T3-E1. Experiments were designed to identify the effects of estrogen on osteoblastic activities associated with both the formation and the resorption of bone. Estrogen treatment coordinately increased DNA content and alkaline phosphatase activity in MC3T3-E1 cells as much as twofold. The stimulatory effect on alkaline phosphatase was stereospecific, dose-dependent between 0.1 and ten nanomolar, and dependent on the time in culture when the hormone was administered. The effect was also persistent, since alkaline phosphatase activity remained elevated for several days after withdrawal of the hormone. Estrogen increased the levels of messenger RNA for alkaline phosphatase and type-I collagen as well, and these effects also persisted after removal of the hormone. The levels of messenger RNA for osteopontin, another bone-matrix protein, were only slightly affected by estrogen. Finally, estrogen inhibited the activation of adenylate cyclase by three osteotropic agents known to stimulate the resorption of bone: parathyroid hormone, prostaglandin E2, and the beta-adrenergic agonist isoproterenol. Thus, estrogen promoted the expression of traits associated with the formation of bone while reducing cellular responsiveness to hormones that may trigger the resorption of bone.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Bone Resorption/metabolism , Estradiol/pharmacology , Estrogens/physiology , Osteoblasts/metabolism , Osteogenesis/physiology , 3T3 Cells/drug effects , 3T3 Cells/metabolism , Adenylyl Cyclases/metabolism , Alkaline Phosphatase/metabolism , Animals , Collagen/biosynthesis , Dinoprostone/antagonists & inhibitors , Isoproterenol/antagonists & inhibitors , Mice , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteogenesis/drug effects , Parathyroid Hormone/antagonists & inhibitors , RNA, Messenger/analysis
20.
J Bone Miner Res ; 8(3): 277-89, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8456584

ABSTRACT

Two sets of clonal cell populations differing in the expression of osteoblastic traits, the rat osteosarcoma cell lines ROS 17/2.8 and ROS 25/1 and the immortalized fetal rat calvarial cell lines RCT-1 and RCT-3, were compared for their ability to attach to a series of extracellular matrix (ECM) constituents in vitro. Both osteoblastic (ROS 17/2.8, RCT-3) and nonosteoblastic (ROS 25/1, RCT-1) cell lines attached in a time- and concentration-dependent manner to plates coated with fibronectin (FN), osteopontin (OP), type I collagen (Col I), type IV collagen (Col IV), and laminin (LN) but only weakly to osteocalcin (OC) and thrombospondin (TSP). In both systems, the osteoblastic and nonosteoblastic clones attached identically to FN. Both ROS 17/2.8 and ROS 25/1 attached to similar molar amounts of substrate with the same preference order: FN > LN > Col I > or = Col IV. Maximal ROS 17/2.8 attachment to OP was > or = Col I but required approximately 2.5 times more substrate. ROS 25/1 attached less effectively than ROS 17/2.8 to most non-FN substrates. RCT-3 cells attached similarly to ROS 17/2.8 except that the preference order for Col I and LN was reversed and attachment to OP was lower than for ROS 17/2.8 RCT-1 cells attached best to Col I rather than FN, and equaled or surpassed RCT-3 in attachment to other non-FN substrates. Thus in these experimental systems, cells expressing an osteoblast-like phenotype exhibited generally similar ECM attachment properties. Their nonosteoblastic counterparts recognized the same spectrum of ECM constituents but differed from the osteoblastic cells and from each other in the effectiveness of their attachment to substrates other than FN.


Subject(s)
Extracellular Matrix/metabolism , Osteoblasts/metabolism , Animals , Cell Adhesion , Cell Line , Collagen/metabolism , Fibronectins/metabolism , Laminin/metabolism , Osteoblasts/physiology , Osteopontin , Protein Binding , Rats , Sialoglycoproteins/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...