Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 276
Filter
1.
J Struct Biol ; 213(4): 107781, 2021 12.
Article in English | MEDLINE | ID: mdl-34411695

ABSTRACT

The interphase region at the base of the growth plate includes blood vessels, cells and mineralized tissues. In this region, cartilage is mineralized and replaced with bone. Blood vessel extremities permeate this space providing nutrients, oxygen and signaling factors. All these different components form a complex intertwined 3D structure. Here we use cryo-FIB SEM to elaborate this 3D structure without removing the water. As it is challenging to image mineralized and unmineralized tissues in a hydrated state, we provide technical details of the parameters used. We obtained two FIB SEM image stacks that show that the blood vessels are in intimate contact not only with cells, but in some locations also with mineralized tissues. There are abundant red blood cells at the extremities of the vessels. We also documented large multinucleated cells in contact with mineralized cartilage and possibly also with bone. We observed membrane bound mineralized particles in these cells, as well as in blood serum, but not in the hypertrophic chondrocytes. We confirm that there is an open pathway from the blood vessel extremities to the mineralizing cartilage. Based on the sparsity of the mineralized particles, we conclude that mainly ions in solution are used for mineralizing cartilage and bone, but these are augmented by the supply of mineralized particles.


Subject(s)
Cartilage/ultrastructure , Cryoelectron Microscopy/methods , Growth Plate/ultrastructure , Imaging, Three-Dimensional/methods , Microscopy, Electron, Scanning/methods , Tibia/ultrastructure , Animals , Basement Membrane/ultrastructure , Blood Vessels/cytology , Blood Vessels/ultrastructure , Bone Development , Calcification, Physiologic , Cartilage/cytology , Cartilage/growth & development , Cell Differentiation , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrocytes/ultrastructure , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Female , Growth Plate/cytology , Growth Plate/growth & development , Mice, Inbred BALB C , Morphogenesis , Tibia/cytology , Tibia/growth & development
2.
Histol Histopathol ; 36(10): 1021-1036, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34137454

ABSTRACT

Hypertrophic chondrocytes are the master regulators of endochondral ossification; however, their ultimate cell fates cells remain largely elusive due to their transient nature. Historically, hypertrophic chondrocytes have been considered as the terminal state of growth plate chondrocytes, which are destined to meet their inevitable demise at the primary spongiosa. Chondrocyte hypertrophy is accompanied by increased organelle synthesis and rapid intracellular water uptake, which serve as the major drivers of longitudinal bone growth. This process is delicately regulated by major signaling pathways and their target genes, including growth hormone (GH), insulin growth factor-1 (IGF-1), indian hedgehog (Ihh), parathyroid hormone-related protein (PTHrP), bone morphogenetic proteins (BMPs), sex determining region Y-box 9 (Sox9), runt-related transcription factors (Runx) and fibroblast growth factor receptors (FGFRs). Hypertrophic chondrocytes orchestrate endochondral ossification by regulating osteogenic-angiogenic and osteogenic-osteoclastic coupling through the production of vascular endothelial growth factor (VEGF), receptor activator of nuclear factor kappa-B ligand (RANKL) and matrix metallopeptidases-9/13 (MMP-9/13). Hypertrophic chondrocytes also indirectly regulate resorption of the cartilaginous extracellular matrix, by controlling formation of a special subtype of osteoclasts termed "chondroclasts". Notably, hypertrophic chondrocytes may possess innate potential for plasticity, reentering the cell cycle and differentiating into osteoblasts and other types of mesenchymal cells in the marrow space. We may be able to harness this unique plasticity for therapeutic purposes, for a variety of skeletal abnormalities and injuries. In this review, we discuss the morphological and molecular properties of hypertrophic chondrocytes, which carry out important functions during skeletal growth and regeneration.


Subject(s)
Chondrocytes/physiology , Chondrocytes/ultrastructure , Growth Plate/physiology , Osteogenesis/physiology , Animals , Cell Size , Chondrogenesis , Growth Plate/cytology , Growth Plate/ultrastructure , Humans , Osteogenesis/genetics
3.
Methods Mol Biol ; 2230: 379-396, 2021.
Article in English | MEDLINE | ID: mdl-33197027

ABSTRACT

Populations of bone marrow stromal cells (BMSCs, also known as bone marrow-derived "mesenchymal stem cells") contain a subset of cells that are able to recapitulate the formation of a bone/marrow organ (skeletal stem cells, SSCs). It is now apparent that cells with similar but not identical properties can be isolated from other skeletal compartments (growth plate, periosteum). The biological properties of BMSCs, and these related stem/progenitor cells, are assessed by a variety of assays, both in vitro and in vivo. Application of these assays in an appropriate fashion provide a great deal of information on the role of BMSCs, and the subset of SSCs, in health and in disease.


Subject(s)
Bone Marrow Cells/ultrastructure , Bone and Bones/ultrastructure , Colony-Forming Units Assay/methods , Mesenchymal Stem Cells/ultrastructure , Animals , Cell Differentiation/genetics , Growth Plate/ultrastructure , Humans
4.
Int J Mol Sci ; 20(15)2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31374825

ABSTRACT

Because Mg-Ca-Zn alloys are biodegradable and obviate secondary implant removal, they are especially beneficial for pediatric patients. We examined the degradation performance of Mg-Ca-Zn alloys depending on the surface modification and investigated the in vivo effects on the growth plate in a skeletally immature rabbit model. Either plasma electrolyte oxidation (PEO)-coated (n = 18) or non-coated (n = 18) Mg-Ca-Zn alloy was inserted at the distal femoral physis. We measured the degradation performance and femoral segment lengths using micro-CT. In addition, we analyzed the histomorphometric and histopathologic characteristics of the growth plate. Although there were no acute, chronic inflammatory reactions in either group, they differed significantly in the tissue reactions to their degradation performance and physeal responses. Compared to non-coated alloys, PEO-coated alloys degraded significantly slowly with diminished hydrogen gas formation. Depending on the degradation rate, large bone bridge formation and premature physeal arrest occurred primarily in the non-coated group, whereas only a small-sized bone bridge formed in the PEO-coated group. This difference ultimately led to significant shortening of the femoral segment in the non-coated group. This study suggests that optimal degradation could be achieved with PEO-coated Mg-Ca-Zn alloys, making them promising and safe biodegradable materials with no growth plate damage.


Subject(s)
Absorbable Implants , Alloys/chemistry , Calcium/chemistry , Growth Plate/physiology , Magnesium/chemistry , Zinc/chemistry , Animals , Bone Nails , Coated Materials, Biocompatible/chemistry , Electrolytes/chemistry , Growth Plate/ultrastructure , Materials Testing , Oxidation-Reduction , Rabbits , Surface Properties
5.
Bioelectrochemistry ; 126: 1-11, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30471483

ABSTRACT

The growth plate is a cartilaginous layer present from the gestation period until the end of puberty where it ossifies joining diaphysis and epiphysis. During this period several endocrine, autocrine, and paracrine processes within the growth plate are carried out by chondrocytes; therefore, a disruption in cellular functions may lead to pathologies affecting bone development. It is known that electric fields impact the growth plate; however, parameters such as stimulation time and electric field intensity are not well documented. Accordingly, this study presents a histomorphometrical framework to assess the effect of electric fields on chondroepiphysis explants. Bones were stimulated with 3.5 and 7 mV/cm, and for each electric field two exposure times were tested for 30 days (30 min and 1 h). Results evidenced that electric fields increased the hypertrophic zones compared with controls. In addition, a stimulation of 3.5 mV/cm applied for 1 h preserved the columnar cell density and its orientation. Moreover, a pre-hypertrophy differentiation in the center of the chondroepiphysis was observed when explants were stimulated during 1 h with both electric fields. These findings allow the understanding of the effect of electrical stimulation over growth plate organization and how the stimulation modifies chondrocytes morphophysiology.


Subject(s)
Chondrocytes/cytology , Electric Stimulation , Growth Plate/growth & development , Animals , Cell Proliferation , Cells, Cultured , Chondrocytes/pathology , Chondrocytes/ultrastructure , Electric Stimulation/instrumentation , Equipment Design , Femur/cytology , Femur/growth & development , Femur/pathology , Femur/ultrastructure , Growth Plate/cytology , Growth Plate/pathology , Growth Plate/ultrastructure , Humerus/cytology , Humerus/growth & development , Humerus/pathology , Humerus/ultrastructure , Hypertrophy , Osteogenesis , Rats , Rats, Wistar
6.
Int J Med Sci ; 15(5): 436-446, 2018.
Article in English | MEDLINE | ID: mdl-29559832

ABSTRACT

Idiopathic scoliosis is one of the most common disabling pathologies of children and adolescents. Etiology and pathogenesis of idiopathic scoliosis remain unknown. To study the etiology of this disease we identified the cells' phenotypes in the vertebral body growth plates in patients with idiopathic scoliosis. Materials and methods: The cells were isolated from vertebral body growth plates of the convex and concave sides of the deformity harvested intraoperatively in 50 patients with scoliosis. Cells were cultured and identified by methods of common morphology, neuromorphology, electron microscopy, immunohistochemistry and PCR analysis. Results: Cultured cells of convex side of deformation were identified as chondroblasts. Cells isolated from the growth plates of the concave side of the deformation showed numerous features of neuro- and glioblasts. These cells formed synapses, contain neurofilaments, and expressed neural and glial proteins. Conclusion: For the first time we demonstrated the presence of cells with neural/glial phenotype in the concave side of the vertebral body growth plate in scoliotic deformity. We hypothesized that neural and glial cells observed in the growth plates of the vertebral bodies represent derivatives of neural crest cells deposited in somites due to alterations in their migratory pathway during embryogenesis. We also propose that ectopic localization of cells derived from neural crest in the growth plate of the vertebral bodies is the main etiological factor of the scoliotic disease.


Subject(s)
Growth Plate/pathology , Neural Crest/pathology , Neuroglia/pathology , Scoliosis/pathology , Adolescent , Child , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrocytes/ultrastructure , Embryonic Development/genetics , Female , Gene Expression Regulation/genetics , Growth Plate/metabolism , Growth Plate/ultrastructure , Humans , Male , Microscopy, Electron, Scanning , Neural Crest/metabolism , Neural Crest/ultrastructure , Neuroglia/metabolism , Scoliosis/etiology , Scoliosis/genetics , Spine/metabolism , Spine/pathology , Spine/ultrastructure
7.
J Bone Miner Res ; 32(10): 2128-2141, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28304100

ABSTRACT

Autophagy is activated during nutritionally depleted or hypoxic conditions to facilitate cell survival. Because growth plate is an avascular and hypoxic tissue, autophagy may have a crucial role during chondrogenesis; however, the functional role and underlying mechanism of autophagy in regulation of growth plate remains elusive. In this study, we generated TamCart Atg7-/- (Atg7cKO) mice to explore the role of autophagy during endochondral ossification. Atg7cKO mice exhibited growth retardation associated with reduced chondrocyte proliferation and differentiation, and increased chondrocyte apoptosis. Meanwhile, we observed that Atg7 ablation mainly induced the PERK-ATF4-CHOP axis of the endoplasmic reticulum (ER) stress response in growth plate chondrocytes. Although Atg7 ablation induced ER stress in growth plate chondrocytes, the addition of phenylbutyric acid (PBA), a chemical chaperone known to attenuate ER stress, partly neutralized such effects of Atg7 ablation on longitudinal bone growth, indicating the causative interaction between autophagy and ER stress in growth plate. Consistent with these findings in vivo, we also observed that Atg7 ablation in cultured chondrocytes resulted in defective autophagy, elevated ER stress, decreased chondrocytes proliferation, impaired expression of col10a1, MMP-13, and VEGFA for chondrocyte differentiation, and increased chondrocyte apoptosis, while such effects were partly nullified by reduction of ER stress with PBA. In addition, Atg7 ablation-mediated impaired chondrocyte function (chondrocyte proliferation, differentiation, and apoptosis) was partly reversed in CHOP-/- cells, indicating the causative role of the PERK-ATF4-CHOP axis of the ER stress response in the action of autophagy deficiency in chondrocytes. In conclusion, our findings indicate that autophagy deficiency may trigger ER stress in growth plate chondrocytes and contribute to growth retardation, thus implicating autophagy as an important regulator during chondrogenesis and providing new insights into the clinical potential of autophagy in cartilage homeostasis. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Activating Transcription Factor 4/metabolism , Autophagy , Cartilage/metabolism , Chondrogenesis , Endoplasmic Reticulum Stress , Transcription Factor CHOP/metabolism , eIF-2 Kinase/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein 7/deficiency , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Cartilage/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/ultrastructure , Chondrogenesis/drug effects , Embryonic Development/drug effects , Endoplasmic Reticulum Stress/drug effects , Femur/drug effects , Femur/growth & development , Gene Deletion , Growth Plate/embryology , Growth Plate/metabolism , Growth Plate/ultrastructure , Mice, Knockout , Organ Specificity , Osteogenesis/drug effects , Phenylbutyrates/pharmacology , Tibia/drug effects , Tibia/growth & development
8.
Eur Cell Mater ; 33: 130-142, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28197989

ABSTRACT

Limitations associated with demineralised bone matrix and other grafting materials have motivated the development of alternative strategies to enhance the repair of large bone defects. The growth plate (GP) of developing limbs contain a plethora of growth factors and matrix cues which contribute to long bone growth, suggesting that biomaterials derived from its extracellular matrix (ECM) may be uniquely suited to promoting bone regeneration. The goal of this study was to generate porous scaffolds from decellularised GP ECM and to evaluate their ability to enhance host mediated bone regeneration following their implantation into critically-sized rat cranial defects. The scaffolds were first assessed by culturing with primary human macrophages, which demonstrated that decellularisation resulted in reduced IL-1ß and IL-8 production. In vitro, GP derived scaffolds were found capable of supporting osteogenesis of mesenchymal stem cells via either an intramembranous or an endochondral pathway, demonstrating the intrinsic osteoinductivity of the biomaterial. Furthermore, upon implantation into cranial defects, GP derived scaffolds were observed to accelerate vessel in-growth, mineralisation and de novo bone formation. These results support the use of decellularised GP ECM as a scaffold for large bone defect regeneration.


Subject(s)
Bone Regeneration , Bone and Bones/pathology , Extracellular Matrix/metabolism , Growth Plate/metabolism , Tissue Scaffolds/chemistry , Wound Healing , Animals , Bone and Bones/diagnostic imaging , Chondrogenesis , Cytokines/biosynthesis , Glycosaminoglycans/metabolism , Growth Plate/ultrastructure , Humans , Macrophages/cytology , Male , Osteogenesis , Phenotype , Porosity , Rats, Inbred F344 , Skull/diagnostic imaging , Skull/pathology , Sus scrofa , X-Ray Microtomography
9.
Dev Dyn ; 246(4): 291-309, 2017 04.
Article in English | MEDLINE | ID: mdl-27987249

ABSTRACT

Autosomal dominant mutations in fibroblast growth factor receptor 3 (FGFR3) cause achondroplasia (Ach), the most common form of dwarfism in humans, and related chondrodysplasia syndromes that include hypochondroplasia (Hch), severe achondroplasia with developmental delay and acanthosis nigricans (SADDAN), and thanatophoric dysplasia (TD). FGFR3 is expressed in chondrocytes and mature osteoblasts where it functions to regulate bone growth. Analysis of the mutations in FGFR3 revealed increased signaling through a combination of mechanisms that include stabilization of the receptor, enhanced dimerization, and enhanced tyrosine kinase activity. Paradoxically, increased FGFR3 signaling profoundly suppresses proliferation and maturation of growth plate chondrocytes resulting in decreased growth plate size, reduced trabecular bone volume, and resulting decreased bone elongation. In this review, we discuss the molecular mechanisms that regulate growth plate chondrocytes, the pathogenesis of Ach, and therapeutic approaches that are being evaluated to improve endochondral bone growth in people with Ach and related conditions. Developmental Dynamics 246:291-309, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Achondroplasia , Receptor, Fibroblast Growth Factor, Type 3/genetics , Signal Transduction/physiology , Achondroplasia/etiology , Achondroplasia/pathology , Achondroplasia/therapy , Animals , Chondrocytes/metabolism , Growth Plate/cytology , Growth Plate/metabolism , Growth Plate/ultrastructure , Humans , Receptor, Fibroblast Growth Factor, Type 3/physiology
10.
Matrix Biol ; 50: 1-15, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26454027

ABSTRACT

The growth plate (GP) is a dynamic tissue driving bone elongation through chondrocyte proliferation, hypertrophy and matrix production. The extracellular matrix (ECM) is the major determinant of GP biomechanical properties and assumed to play a pivotal role for chondrocyte geometry and arrangement, thereby guiding proper growth plate morphogenesis and bone elongation. To elucidate the relationship between morphology and biomechanics during cartilage morphogenesis, we have investigated age-dependent structural and elastic properties of the proliferative zone of the murine GP by atomic force microscopy (AFM) from the embryonic stage to adulthood. We observed a progressive cell flattening and arrangement into columns from embryonic day 13.5 until postnatal week 2, correlating with an increasing collagen density and ECM stiffness, followed by a nearly constant cell shape, collagen density and ECM stiffness from week 2 to 4 months. At all ages, we found marked differences in the density and organization of the collagen network between the intracolumnar matrix, and the intercolumnar matrix, associated with a roughly two-fold higher stiffness of the intracolumnar matrix compared to the intercolumnar matrix. This difference in local ECM stiffness may force the cells to arrange in a columnar structure upon cell division and drive bone elongation during embryonic and juvenile development.


Subject(s)
Cartilage, Articular/growth & development , Growth Plate/physiology , Growth Plate/ultrastructure , Animals , Biomechanical Phenomena , Cartilage, Articular/physiology , Cell Proliferation , Extracellular Matrix/physiology , Growth Plate/growth & development , Mice , Microscopy, Atomic Force , Stress, Mechanical
11.
Nat Commun ; 6: 6798, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25865282

ABSTRACT

The diverse morphology of vertebrate skeletal system is genetically controlled, yet the means by which cells shape the skeleton remains to be fully illuminated. Here we perform quantitative analyses of cell behaviours in the growth plate cartilage, the template for long bone formation, to gain insights into this process. Using a robust avian embryonic organ culture, we employ time-lapse two-photon laser scanning microscopy to observe proliferative cells' behaviours during cartilage growth, resulting in cellular trajectories with a spreading displacement mainly along the tissue elongation axis. We build a novel software toolkit of quantitative methods to segregate the contributions of various cellular processes to the cellular trajectories. We find that convergent-extension, mitotic cell division, and daughter cell rearrangement do not contribute significantly to the observed growth process; instead, extracellular matrix deposition and cell volume enlargement are the key contributors to embryonic cartilage elongation.


Subject(s)
Cartilage/ultrastructure , Chondrocytes/ultrastructure , Fibroblasts/ultrastructure , Growth Plate/ultrastructure , Metacarpal Bones/ultrastructure , Osteogenesis/physiology , Animals , Cartilage/embryology , Cartilage/metabolism , Cell Division , Cell Movement , Cell Size , Chick Embryo , Chondrocytes/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Gene Expression , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Growth Plate/embryology , Growth Plate/metabolism , Metacarpal Bones/embryology , Metacarpal Bones/metabolism , Microscopy, Confocal , Organ Culture Techniques , Photons , Retroviridae/genetics , Time-Lapse Imaging
12.
Mol Genet Metab ; 114(2): 195-202, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24953405

ABSTRACT

We treated mucopolysaccharidosis IVA (MPS IVA) mice to assess the effects of long-term enzyme replacement therapy (ERT) initiated at birth, since adult mice treated by ERT showed little improvement in bone pathology [1]. To conduct ERT in newborn mice, we used recombinant human N-acetylgalactosamine-6-sulfate sulfatase (GALNS) produced in a CHO cell line. First, to observe the tissue distribution pattern, a dose of 250units/g body weight was administered intravenously in MPS IVA mice at day 2 or 3. The infused enzyme was primarily recovered in the liver and spleen, with detectable activity in the bone and brain. Second, newborn ERT was conducted after a tissue distribution study. The first injection of newborn ERT was performed intravenously, the second to fourth weekly injections were intraperitoneal, and the remaining injections from 5th to 14th weeks were intravenous into the tail vein. MPS IVA mice treated with GALNS showed clearance of lysosomal storage in the liver and spleen, and sinus lining cells in bone marrow. The column structure of the growth plate was organized better than that in adult mice treated with ERT; however, hyaline and fibrous cartilage cells in the femur, spine, ligaments, discs, synovium, and periosteum still had storage materials to some extent. Heart valves were refractory to the treatment. Levels of serum keratan sulfate were kept normal in newborn ERT mice. In conclusion, the enzyme, which enters the cartilage before the cartilage cell layer becomes mature, prevents disorganization of column structure. Early treatment from birth leads to partial remission of bone pathology in MPS IVA mice.


Subject(s)
Bone Diseases/drug therapy , Chondroitinsulfatases/therapeutic use , Enzyme Replacement Therapy , Mucopolysaccharidosis IV/drug therapy , Administration, Intravenous , Animals , Animals, Newborn , Bone Diseases/pathology , CHO Cells , Cartilage/drug effects , Cartilage/ultrastructure , Chondrocytes/drug effects , Chondrocytes/ultrastructure , Chondroitinsulfatases/administration & dosage , Chondroitinsulfatases/genetics , Chondroitinsulfatases/pharmacokinetics , Cricetulus , Disease Models, Animal , Growth Plate/drug effects , Growth Plate/ultrastructure , Keratan Sulfate/blood , Liver/drug effects , Mice , Mice, Knockout , Mucopolysaccharidosis IV/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Spleen/drug effects , Tissue Distribution/drug effects
13.
BMC Musculoskelet Disord ; 15: 347, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25319082

ABSTRACT

BACKGROUND: The cn/cn dwarf mouse is caused by a loss-of-function mutation in the natriuretic peptide receptor 2 (NPR-2) gene which helps positively regulate endochondral longitudinal bone growth. The gene mutation corresponds to that in the human skeletal dysplasia Acromesomelic Dysplasia Maroteaux type (AMDM). This study assesses histomorphometric, ultrastructural and radiographic correlates of the growth abnormality. METHODS: Ten litters of cn/cn and cn/+littermates at ages ranging from 2.5 to 6.5 weeks were studied by skeletal radiographs, histomorphometry and physeal ultrastructure. Skeletal radiographs were done on 2 cn/cn and 2 cn/+littermates at 5 weeks of age. Humeral, femoral, and tibial lengths were measured from 34 intact bones (17 cn/cn, 17 cn/+) at 2.5 to 6.5 weeks. Growth plate histomorphometry in 50 bones (26 cn/cn and 24 cn/+) determined the hypertrophic zone/entire physeal cartilage ratios in 204 sections (87 cn/+, 117 cn/cn) at 3 time periods (2.5-3, 4-4.5, and 6-6.5 weeks). Electron microscopy assessed 6 cn/cn and 6 cn/+age and site-matched physeal cartilage. RESULTS: Cn/cn mice were two thirds the size of the cn/+. Cn/cn bones were normal in shape or only minimally deformed except for the radius with mid-diaphyseal bowing. Length ratios of cn/cn humeri, femurs, and tibias were a mean of 0.65 (± 0.03, n = 34, 17 ratios) compared to cn/+bones. The main physeal abnormality was a markedly shortened hypertrophic zone with the ratio of hypertrophic zone to entire physis 0.17 (± 0.063) in the cn/cn and 0.30 (± 0.052) in the cn/+mice. Ratio assessments were similar comparing humeral, femoral, and tibial growth plates as were ratios from each of the 3 time periods. Ultrastructural assessments from the resting zone to the lower hypertrophic zone-metaphyseal junction showed no specific individual cell abnormalities in cn/cn compared to cn/+physes. CONCLUSIONS: The disorder causes a shortened physeal hypertrophic zone but normal ultrastructure of cn/cn chondrocytes points to abnormality primarily affecting the hypertrophic zone rather than a structural cell or matrix synthesis problem.


Subject(s)
Bone Diseases, Developmental/diagnostic imaging , Bone Diseases, Developmental/pathology , Bone and Bones/pathology , Bone and Bones/ultrastructure , Animals , Body Weight , Bone and Bones/diagnostic imaging , Chondrocytes/ultrastructure , Disease Models, Animal , Epiphyses/pathology , Epiphyses/ultrastructure , Femur/diagnostic imaging , Femur/pathology , Femur/ultrastructure , Growth Plate/pathology , Growth Plate/ultrastructure , Humans , Humerus/diagnostic imaging , Humerus/pathology , Humerus/ultrastructure , Mice , Mice, Mutant Strains , Radiography , Tibia/diagnostic imaging , Tibia/pathology , Tibia/ultrastructure
14.
J Morphol ; 275(11): 1226-37, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24895174

ABSTRACT

To date, little is known about the structure of the cells and the fibrillar matrix of the globuli ossei, globular structures showing histochemical properties of an osseous tissue, sometimes found in the resorption front of the hypertrophied cartilage in many tetrapods, and easily observed in the long bones of the Urodele Pleurodeles waltl. Here, we present the results obtained from the appendicular long bones of metamorphosed juveniles and subadults using histological and histochemical methods and transmission electron microscopy. The distal part of the cone-shaped cartilage contains a heterogeneous cell population composed of the typical "light" hypertrophic chondrocytes and scarce "dark" hypertrophic chondrocytes. The "dark" chondrocytes display ultrastructural characteristics suggesting that they probably undergo degeneration through chondroptosis. However, in the hypertrophic, calcified cartilage close to the erosion front by the marrow, several noninvaded chondrocytic lacunae retained cells that do not show any morphological characteristics of degeneration and that cannot be identified as regular chondrocytes or osteocytes. These modified chondrocytes that have lost their regular morphology, appear to be active in the terminal cartilage and synthesize collagen fibrils of a peculiar diameter intermediate between the Type I collagen found in bone and the Type II collagen characteristic of cartilage. It is suggested that the local occurrence of globuli ossei is linked to a low rate of longitudinal growth as is the case in the long bones of postmetamorphic urodeles.


Subject(s)
Bone and Bones/anatomy & histology , Chondrocytes/ultrastructure , Growth Plate/ultrastructure , Hyaline Cartilage/ultrastructure , Osteocytes/ultrastructure , Pleurodeles/anatomy & histology , Animals , Bone Development , Cell Differentiation , Extracellular Matrix , Extremities/anatomy & histology , Hypertrophy , Microscopy, Electron, Transmission
15.
J Anat ; 223(6): 641-50, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24117386

ABSTRACT

Overgrowth of limbs and spinal deformities are typical clinical manifestations of Marfan syndrome (MFS) and congenital contractural arachnodactyly (CCA), caused by mutations of the genes encoding fibrillin-1 (FBN1) and fibrillin-2 (FBN2), respectively. FBN1 mutations are also associated with acromicric (AD) and geleophysic dysplasias (GD), and with Weill-Marchesani syndrome (WMS), which is characterised by short stature. The mechanisms leading to such abnormal skeletal growth and the involvement of the fibrillins are not understood. Postnatal longitudinal bone growth mainly occurs in the epiphyseal growth plate. Here we investigated the organisation of fibrillin microfibrils in the growth plate of the long bone and vertebra immunohistochemically. Fibrillin-1 was dual-immunostained with elastin, with fibrillin-2 or with collagen X. We report that fibrillin microfibrils are distributed throughout all regions of the growth plate, and that fibrillin-1 and fibrillin-2 were differentially organised. Fibrillin-1 was more abundant in the extracellular matrix of the resting and proliferative zones of the growth plate than in the hypertrophic zone. More fibrillin-2 was found in the calcified region than in the other regions. No elastin fibres were observed in either the proliferative or hypertrophic zones. This study indicates that, as fibrillin microfibrils are involved in growth factor binding and may play a mechanical role, they could be directly involved in regulating bone growth. Hence, mutations of the fibrillins could affect their functional role in growth and lead to the growth disorders seen in patients with MFS, CCA, AD, GD and WMS.


Subject(s)
Growth Plate/chemistry , Metacarpal Bones/chemistry , Microfibrils/chemistry , Microfilament Proteins/analysis , Spine/chemistry , Animals , Cattle , Collagen/analysis , Elastin/analysis , Fibrillins , Growth Plate/ultrastructure , Immunohistochemistry , Metacarpal Bones/ultrastructure , Microfilament Proteins/physiology , Spine/ultrastructure
16.
Int J Biol Sci ; 9(9): 895-906, 2013.
Article in English | MEDLINE | ID: mdl-24163588

ABSTRACT

Bone morphogenic proteins (BMPs) are critical for both chondrogenesis and osteogenesis. Previous studies reported that embryos deficient in Bmp receptor (Bmpr)1a or Bmpr1b in cartilage display subtle skeletal defects; however, double mutant embryos develop severe skeletal defects, suggesting a functional redundancy that is essential for early chondrogenesis. In this study, we examined the postnatal role of Bmpr1a in cartilage. In the Bmpr1a conditional knockout (cKO, a cross between Bmpr1a flox and aggrecan-CreER (T2) induced by a one-time-tamoxifen injection at birth and harvested at ages of 2, 4, 8 and 20 weeks), there was essentially no long bone growth with little expression of cartilage markers such as SOX9, IHH and glycoproteins. Unexpectedly, the null growth plate was replaced by bone-like tissues, supporting the notions that the progenitor cells in the growth plate, which normally form cartilage, can form other tissues such as bone and fibrous; and that BMPR1A determines the cell fate. A working hypothesis is proposed to explain the vital role of BMPR1A in postnatal chondrogenesis.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/physiology , Animals , Bone Development/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cartilage/growth & development , Cartilage/metabolism , Cartilage/ultrastructure , Cell Differentiation , Chondrogenesis/genetics , Gene Expression Regulation, Developmental , Growth Plate/cytology , Growth Plate/metabolism , Growth Plate/ultrastructure , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Tamoxifen
17.
Biomech Model Mechanobiol ; 12(1): 67-78, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22446833

ABSTRACT

Mechanical environment is one of the regulating factors involved in the process of longitudinal bone growth. Non-physiological compressive loading can lead to infantile and juvenile musculoskeletal deformities particularly during growth spurt. We hypothesized that tissue mechanical behavior in sub-regions (reserve, proliferative and hypertrophic zones) of the growth plate is related to its collagen and proteoglycan content as well as its collagen fiber orientation. To characterize the strain distribution through growth plate thickness and to evaluate biochemical content and collagen fiber organization of the three histological zones of growth plate tissue. Distal ulnar growth plate samples (N = 29) from 4-week old pigs were analyzed histologically for collagen fiber organization (N = 7) or average zonal thickness (N = 8), or trimmed into the three average zones, based on the estimated thickness of each histological zone, for biochemical analysis of water, collagen and glycosaminoglycan content (N = 7). Other samples (N = 7) were tested in semi-confined compression under 10% compressive strain. Digital images of the fluorescently labeled nuclei were concomitantly acquired by confocal microscopy before loading and after tissue relaxation. Strain fields were subsequently calculated using a custom-designed 2D digital image correlation algorithm. Depth-dependent compressive strain patterns and collagen content were observed. The proliferative and hypertrophic zone developed the highest axial and transverse strains, respectively, under compression compared to the reserve zone, in which the lowest axial and transverse strains arose. The collagen content per wet mass was significantly lower in the proliferative and hypertrophic zones compared to the reserve zone, and all three zones had similar glycosaminoglycan and water content.Polarized light microscopy showed that collagen fibers were mainly organized horizontally in the reserve zone and vertically aligned with the growth direction in the proliferative and hypertrophic zones. Higher strains were developed in growth plate areas (proliferative and hypertrophic) composed of lower collagen content and of vertical collagen fiber organization. The stiffer reserve zone, with its higher collagen content and collagen fibers oriented to restrain lateral expansion under compression, could play a greater role of mechanical support compared to the proliferative and hypertrophic zones, which could be more susceptible to be involved in an abnormal growth process.


Subject(s)
Extracellular Matrix/physiology , Fibrillar Collagens/physiology , Growth Plate/physiology , Mechanotransduction, Cellular/physiology , Models, Biological , Ulna/physiology , Animals , Compressive Strength/physiology , Computer Simulation , Elastic Modulus/physiology , Extracellular Matrix/ultrastructure , Fibrillar Collagens/ultrastructure , Growth Plate/ultrastructure , In Vitro Techniques , Stress, Mechanical , Swine , Ulna/ultrastructure
18.
J Theor Biol ; 285(1): 147-55, 2011 Sep 21.
Article in English | MEDLINE | ID: mdl-21723296

ABSTRACT

The research investigates the role of the immotile chondrocytic primary cilium in the growth plate. This study was motivated by (i) the recent evidence of the mechano-sensorial function of the primary cilium in kidney tubule epithelial cells and (ii) the distinct three-dimensional orientation patterns that the chondrocytic primary cilium forms in articular cartilage in the presence or the absence of loading. For our investigation, we used the Smad1/5(CKO) mutant mouse, whose disorganized growth plate is due to the conditional deletion of Smad 1 and 5 proteins that also affect the so-called Indian Hedgehog pathway, whose physical and functional topography has been shown to be partially controlled by the primary cilium. Fluorescence and confocal microscopy on stained sections visualized ciliated chondrocytes. Morphometric data regarding position, orientation and eccentricity of chondrocytes, and ciliary localization on cell membrane, length and orientation, were collected and reconstructed from images. We established that both localization and orientation of the cilium are definite, and differently so, in the Smad1/5(CKO) and control mice. The orientation of the primary cilium, relative to the major axis of the chondrocyte, clusters at 80° with respect to the anterior-posterior direction for the Smad1/5(CKO) mice, showing loss of the additional clustering present in the control mice at 10°. We therefore hypothesized that the clustering at 10° contains information of columnar organization. To test our hypothesis, we prepared a mathematical model of relative positioning of the proliferative chondrocytic population based on ciliary orientation. Our model belongs to the category of "interactive particle system models for self-organization with birth". The model qualitatively reproduced the experimentally observed chondrocytic arrangements in growth plate of each of the Smad1/5(CKO) and control mice. Our mathematically predicted cell division process will need to be observed experimentally to advance the identification of ciliary function in the growth plate.


Subject(s)
Chondrocytes/ultrastructure , Cilia/ultrastructure , Growth Plate/ultrastructure , Models, Biological , Animals , Cell Division/physiology , Chondrocytes/physiology , Cilia/physiology , Growth Plate/physiology , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , Orientation , Smad1 Protein/deficiency , Smad1 Protein/physiology , Smad5 Protein/deficiency , Smad5 Protein/physiology
19.
J Pathol ; 224(2): 160-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21506131

ABSTRACT

Proteoglycans are secreted into the extracellular matrix of virtually all cell types and function in several cellular processes. They consist of a core protein onto which glycosaminoglycans (e.g., heparan or chondroitin sulphates), are attached. Proteoglycans are important modulators of gradient formation and signal transduction. Impaired biosynthesis of heparan sulphate glycosaminoglycans causes osteochondroma, the most common bone tumour to occur during adolescence. Cytochemical staining with positively charged dyes (e.g., polyethyleneimine-PEI) allows, visualisation of proteoglycans and provides a detailed description of how proteoglycans are distributed throughout the cartilage matrix. PEI staining was studied by electron and reflection contrast microscopy in human growth plates, osteochondromas and five different proteoglycan-deficient zebrafish mutants displaying one of the following skeletal phenotypes: dackel (dak/ext2), lacking heparan sulphate and identified as a model for human multiple osteochondromas; hi307 (ß3gat3), deficient for most glycosaminoglycans; pinscher (pic/slc35b2), presenting with defective sulphation of glycosaminoglycans; hi954 (uxs1), lacking most glycosaminoglycans; and knypek (kny/gpc4), missing the protein core of the glypican-4 proteoglycan. The panel of genetically well-characterized proteoglycan-deficient zebrafish mutants serves as a convincing and comprehensive study model to investigate proteoglycan distribution and the relation of this distribution to the model mutation status. They also provide insight into the distributions and gradients that can be expected in the human homologue. Human growth plate, wild-type zebrafish and fish mutants with mild proteoglycan defects (hi307 and kny) displayed proteoglycans distributed in a gradient throughout the matrix. Although the mutants pic and hi954, which had severely impaired proteoglycan biosynthesis, showed no PEI staining, dak mutants demonstrated reduced PEI staining and no gradient formation. Most chondrocytes from human osteochondromas showed normal PEI staining. However, approximately 10% of tumour chondrocytes were similar to those found in the dak mutant (e.g., lack of PEI gradients). The cells in the reduced PEI-stained areas are likely associated with loss-of-function mutations in the EXT genes, and they might contribute to tumour initiation by disrupting the gradients.


Subject(s)
Bone Neoplasms/metabolism , Growth Plate/metabolism , Osteochondroma/metabolism , Proteoglycans/metabolism , Adolescent , Adult , Animals , Bone Neoplasms/ultrastructure , Chondrocytes/metabolism , Disease Models, Animal , Growth Plate/ultrastructure , Humans , Microscopy, Electron , Microscopy, Phase-Contrast , Mutation , N-Acetylglucosaminyltransferases/genetics , Neoplasm Proteins/metabolism , Osteochondroma/ultrastructure , Proteoglycans/deficiency , Zebrafish
20.
Acta Orthop ; 81(3): 331-6, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20443746

ABSTRACT

BACKGROUND AND PURPOSE: Several different theories have been proposed to explain the pathogenesis of slipped capital femoral epiphysis (SCFE). Using transmission electron microscopy (TEM), we carried out an ultrastructural study of core biopsy specimens of the physis at various stages of the disease. METHODS: Core biopsies were performed in 6 patients with different forms of SCFE during the first operation, and in 3 of them when removing the osteosynthesis material before physeal closure. The specimens were prepared for TEM examination. RESULTS: In 6 specimens obtained at first surgery, a marked distortion of the physeal architecture was observed. In 2 of the 3 specimens obtained at removal of the osteosynthesis material, the physis showed a more normal organization. INTERPRETATION: The improvement of the pathological alterations observed in the 2 cases after surgical intervention leads us to consider the possibility that when the growth plate is stabilized directly by pinning or indirectly by creating more optimal loading conditions with an intertrochanteric osteotomy, the morpho-functional characteristics of the physis can be restored and its growth process may resume.


Subject(s)
Epiphyses, Slipped/pathology , Femur , Adolescent , Biopsy , Child , Epiphyses, Slipped/etiology , Epiphyses, Slipped/surgery , Female , Femur/surgery , Growth Plate/ultrastructure , Humans , Male , Microscopy, Electron, Transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...