Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Bone Res ; 12(1): 12, 2024 02 23.
Article in English | MEDLINE | ID: mdl-38395992

ABSTRACT

Piezo proteins are mechanically activated ion channels, which are required for mechanosensing functions in a variety of cell types. While we and others have previously demonstrated that the expression of Piezo1 in osteoblast lineage cells is essential for bone-anabolic processes, there was only suggestive evidence indicating a role of Piezo1 and/or Piezo2 in cartilage. Here we addressed the question if and how chondrocyte expression of the mechanosensitive proteins Piezo1 or Piezo2 controls physiological endochondral ossification and pathological osteoarthritis (OA) development. Mice with chondrocyte-specific inactivation of Piezo1 (Piezo1Col2a1Cre), but not of Piezo2, developed a near absence of trabecular bone below the chondrogenic growth plate postnatally. Moreover, all Piezo1Col2a1Cre animals displayed multiple fractures of rib bones at 7 days of age, which were located close to the growth plates. While skeletal growth was only mildly affected in these mice, OA pathologies were markedly less pronounced compared to littermate controls at 60 weeks of age. Likewise, when OA was induced by anterior cruciate ligament transection, only the chondrocyte inactivation of Piezo1, not of Piezo2, resulted in attenuated articular cartilage degeneration. Importantly, osteophyte formation and maturation were also reduced in Piezo1Col2a1Cre mice. We further observed increased Piezo1 protein abundance in cartilaginous zones of human osteophytes. Finally, we identified Ptgs2 and Ccn2 as potentially relevant Piezo1 downstream genes in chondrocytes. Collectively, our data do not only demonstrate that Piezo1 is a critical regulator of physiological and pathological endochondral ossification processes, but also suggest that Piezo1 antagonists may be established as a novel approach to limit osteophyte formation in OA.


Subject(s)
Cartilage, Articular , Osteoarthritis , Osteophyte , Animals , Humans , Mice , Cartilage, Articular/pathology , Chondrocytes , Ion Channels/genetics , Osteoarthritis/genetics , Osteogenesis/genetics , Osteophyte/metabolism
2.
Front Immunol ; 10: 2182, 2019.
Article in English | MEDLINE | ID: mdl-31572390

ABSTRACT

The bone matrix is constantly remodeled by bone-resorbing osteoclasts and bone-forming osteoblasts. These two cell types are fundamentally different in terms of progenitor cells, mode of action and regulation by specific molecules, acting either systemically or locally. Importantly, there is increasing evidence for an impact of cell types or molecules of the adaptive and innate immune system on bone remodeling. Understanding these influences is the major goal of a novel research area termed osteoimmunology, which is of key relevance in the context of inflammation-induced bone loss, skeletal metastases, and diseases of impaired bone remodeling, such as osteoporosis. This review article aims at summarizing the current knowledge on one particular aspect of osteoimmunology, namely the impact of chemokines on skeletal cells in order to regulate bone remodeling under physiological and pathological conditions. Chemokines have key roles in the adaptive immune system by controlling migration, localization, and function of immune cells during inflammation. The vast majority of chemokines are divided into two subgroups based on the pattern of cysteine residues. More specifically, there are 27 known C-C-chemokines, binding to 10 different C-C receptors, and 17 known C-X-C-chemokines binding to seven different C-X-C receptors. Three additional chemokines do not fall into this category, and only one of them, i.e., CX3CL1, has been shown to influence bone remodeling cell types. There is a large amount of published studies demonstrating specific effects of certain chemokines on differentiation and function of osteoclasts and/or osteoblasts. Chemokine signaling by skeletal cells or by other cells of the bone marrow niche regulates bone formation and resorption through autocrine and paracrine mechanisms. In vivo evidence from mouse deficiency models strongly supports the role of certain chemokine signaling pathways in bone remodeling. We will summarize these data in the present review with a special focus on the most established subsets of chemokines. In combination with the other review articles of this issue, the knowledge presented here confirms that there is a physiologically relevant crosstalk between the innate immune system and bone remodeling cell types, whose molecular understanding is of high clinical relevance.


Subject(s)
Bone Resorption , Chemokines/immunology , Osteoblasts , Osteoclasts , Osteogenesis/immunology , Signal Transduction/immunology , Animals , Bone Resorption/immunology , Bone Resorption/pathology , Humans , Mice , Osteoblasts/immunology , Osteoblasts/pathology , Osteoclasts/immunology , Osteoclasts/pathology
3.
PLoS One ; 14(7): e0219734, 2019.
Article in English | MEDLINE | ID: mdl-31314788

ABSTRACT

Albeit osteoporosis is one of the most prevalent disorders in the aged population, treatment options stimulating the activity of bone-forming osteoblasts are still limited. We and others have previously identified sphingosine-1-phosphate (S1P) as a bone remodeling coupling factor, which is released by bone-resorbing osteoclasts to stimulate bone formation. Moreover, S1pr3, encoding one of the five known S1P receptors (S1P3), was found differentially expressed in osteoblasts, and S1P3 deficiency corrected the moderate high bone mass phenotype of a mouse model (deficient for the calcitonin receptor) with increased S1P release from osteoclasts. In the present study we addressed the question, if S1P3 deficiency would also influence the skeletal phenotype of mice lacking S1P-lyase (encoded by Sgpl1), which display markedly increased S1P levels due to insufficient degradation. Consistent with previous reports, the majority of Sgpl1-deficient mice died before or shortly after weaning, and this lethality was not influenced by additional S1P3 deficiency. At 3 weeks of age, Sgpl1-deficient mice displayed increased trabecular bone mass, which was associated with enhanced osteoclastogenesis and bone resorption, but also with increased bone formation. Most importantly however, none of the skeletal parameters assessed by µCT, histomorphometry and serum analyses were significantly influenced by additional S1P3 deficiency. Taken together, our findings fully support the concept that S1P is a potent osteoanabolic molecule, although S1P3 is not the sole receptor mediating this influence. Since S1P receptors are considered excellent drug targets, it is now required to screen for the impact of other family members on bone formation.


Subject(s)
Aldehyde-Lyases/genetics , Bone and Bones/enzymology , Sphingosine-1-Phosphate Receptors/genetics , Alleles , Animals , Bone Remodeling , Bone and Bones/diagnostic imaging , Female , Male , Mice , Mice, Transgenic , Osteoblasts/enzymology , Osteoclasts/enzymology , Phenotype , X-Ray Microtomography
4.
PLoS One ; 12(10): e0187030, 2017.
Article in English | MEDLINE | ID: mdl-29088242

ABSTRACT

Fetuin-A / α2-Heremans-Schmid-glycoprotein (gene name Ahsg) is a systemic inhibitor of ectopic calcification. Due to its high affinity for calcium phosphate, fetuin-A is highly abundant in mineralized bone matrix. Foreshortened femora in fetuin-A-deficient Ahsg-/- mice indicated a role for fetuin-A in bone formation. We studied early postnatal bone development in fetuin-A-deficient mice and discovered that femora from Ahsg-/- mice exhibited severely displaced distal epiphyses and deformed growth plates, similar to the human disease slipped capital femoral epiphysis (SCFE). The growth plate slippage occurred in 70% of Ahsg-/- mice of both sexes around three weeks postnatal. At this time point, mice weaned and rapidly gained weight and mobility. Epiphysis slippage never occurred in wildtype and heterozygous Ahsg+/- mice. Homozygous fetuin-A-deficient Ahsg-/- mice and, to a lesser degree, heterozygous Ahsg+/- mice showed lesions separating the proliferative zone from the hypertrophic zone of the growth plate. The hypertrophic growth plate cartilage in long bones from Ahsg-/- mice was significantly elongated and V-shaped until three weeks of age and thus prior to the slippage. Genome-wide transcriptome analysis of laser-dissected distal femoral growth plates from 13-day-old Ahsg-/- mice revealed a JAK-STAT-mediated inflammatory response including a 550-fold induction of the chemokine Cxcl9. At this stage, vascularization of the elongated growth plates was impaired, which was visualized by immunofluorescence staining. Thus, fetuin-A-deficient mice may serve as a rodent model of growth plate pathologies including SCFE and inflammatory cartilage degradation.


Subject(s)
Bone Diseases, Developmental/genetics , Epiphyses, Slipped/genetics , Femur/abnormalities , Hindlimb/abnormalities , alpha-2-HS-Glycoprotein/genetics , Animals , Female , Fluorescent Antibody Technique , Gene Expression Profiling/methods , Growth Plate/abnormalities , Male , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Weaning , alpha-2-HS-Glycoprotein/deficiency
5.
Nat Commun ; 4: 1507, 2013.
Article in English | MEDLINE | ID: mdl-23422675

ABSTRACT

Despite its importance in many industrial, geological and biological processes, the mechanism of crystallization from supersaturated solutions remains a matter of debate. Recent discoveries show that in many solution systems nanometre-sized structural units are already present before nucleation. Still little is known about the structure and role of these so-called pre-nucleation clusters. Here we present a combination of in situ investigations, which show that for the crystallization of calcium phosphate these nanometre-sized units are in fact calcium triphosphate complexes. Under conditions in which apatite forms from an amorphous calcium phosphate precursor, these complexes aggregate and take up an extra calcium ion to form amorphous calcium phosphate, which is a fractal of Ca(2)(HPO(4))(3)(2-) clusters. The calcium triphosphate complex also forms the basis of the crystal structure of octacalcium phosphate and apatite. Finally, we demonstrate how the existence of these complexes lowers the energy barrier to nucleation and unites classical and non-classical nucleation theories.


Subject(s)
Biomimetic Materials/chemistry , Calcium Phosphates/chemistry , Models, Chemical , Animals , Calcium/analysis , Cattle , Collagen/chemistry , Cryoelectron Microscopy , Durapatite/chemistry , Hydrogen-Ion Concentration , Ions , Kinetics , Microscopy, Atomic Force , Models, Molecular , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Particle Size , Polymers/chemistry , Spectroscopy, Fourier Transform Infrared , Static Electricity , Synchrotrons , Thermodynamics , X-Ray Diffraction
6.
Nat Mater ; 9(12): 1004-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20972429

ABSTRACT

Bone is a composite material in which collagen fibrils form a scaffold for a highly organized arrangement of uniaxially oriented apatite crystals. In the periodic 67 nm cross-striated pattern of the collagen fibril, the less dense 40-nm-long gap zone has been implicated as the place where apatite crystals nucleate from an amorphous phase, and subsequently grow. This process is believed to be directed by highly acidic non-collagenous proteins; however, the role of the collagen matrix during bone apatite mineralization remains unknown. Here, combining nanometre-scale resolution cryogenic transmission electron microscopy and cryogenic electron tomography with molecular modelling, we show that collagen functions in synergy with inhibitors of hydroxyapatite nucleation to actively control mineralization. The positive net charge close to the C-terminal end of the collagen molecules promotes the infiltration of the fibrils with amorphous calcium phosphate (ACP). Furthermore, the clusters of charged amino acids, both in gap and overlap regions, form nucleation sites controlling the conversion of ACP into a parallel array of oriented apatite crystals. We developed a model describing the mechanisms through which the structure, supramolecular assembly and charge distribution of collagen can control mineralization in the presence of inhibitors of hydroxyapatite nucleation.


Subject(s)
Apatites/metabolism , Collagen Type I/metabolism , Durapatite/antagonists & inhibitors , Animals , Bone and Bones/metabolism , Calcium Phosphates/metabolism , Collagen Type I/chemistry , Cryoelectron Microscopy , Cryopreservation , Electron Microscope Tomography , Extracellular Matrix/metabolism , Horses , Light , Models, Molecular , Nanoparticles/chemistry , Osteogenesis , Peptides/metabolism , Scattering, Radiation , Spectrometry, X-Ray Emission , Staining and Labeling/methods , Surface Properties , Tendons/chemistry
7.
Chem Commun (Camb) ; 46(10): 1703-5, 2010 Mar 14.
Article in English | MEDLINE | ID: mdl-20177622

ABSTRACT

Chitosan (CS) can mediate the formation of spherical, tabulate, and unique starfruit-like silica in the presence of phosphate ions (Pi). CryoTEM and cryoET were used to examine the CS aggregates in the hydrated state. 3D starfruit-like CS/Pi aggregates were reconstructed, which unambiguously confirmed the templating effect of CS/Pi in biomimetic silicification.


Subject(s)
Biomimetics/methods , Chitosan/chemistry , Electron Microscope Tomography , Silicon Dioxide/chemistry , Carbohydrate Conformation , Models, Molecular , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...