Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.888
Filter
1.
Int J Mol Sci ; 25(8)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38673812

ABSTRACT

Here, we report on the development of a cost-effective, well-characterized three-dimensional (3D) model of bone homeostasis derived from commonly available stocks of immortalized murine cell lines and laboratory reagents. This 3D murine-cell-derived bone organoid model (3D-mcBOM) is adaptable to a range of contexts and can be used in conjunction with surrogates of osteoblast and osteoclast function to study cellular and molecular mechanisms that affect bone homeostasis in vitro or to augment in vivo models of physiology or disease. The 3D-mcBOM was established using a pre-osteoblast murine cell line, which was seeded into a hydrogel extracellular matrix (ECM) and differentiated into functional osteoblasts (OBs). The OBs mineralized the hydrogel ECM, leading to the deposition and consolidation of hydroxyapatite into bone-like organoids. Fourier-transform infrared (FTIR) spectroscopy confirmed that the mineralized matrix formed in the 3D-mcBOM was bone. The histological staining of 3D-mcBOM samples indicated a consistent rate of ECM mineralization. Type I collagen C-telopeptide (CTX1) analysis was used to evaluate the dynamics of OC differentiation and activity. Reliable 3D models of bone formation and homeostasis align with current ethical trends to reduce the use of animal models. This functional model of bone homeostasis provides a cost-effective model system using immortalized cell lines and easily procured supplemental compounds, which can be assessed by measuring surrogates of OB and OC function to study the effects of various stimuli in future experimental evaluations of bone homeostasis.


Subject(s)
Cell Differentiation , Extracellular Matrix , Organoids , Osteoblasts , Osteogenesis , Animals , Mice , Organoids/cytology , Organoids/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Extracellular Matrix/metabolism , Bone and Bones/cytology , Bone and Bones/metabolism , Cell Line , Collagen Type I/metabolism , Hydrogels/chemistry , Calcification, Physiologic , Cell Culture Techniques, Three Dimensional/methods , Models, Biological
2.
Stem Cell Rev Rep ; 20(4): 1106-1120, 2024 May.
Article in English | MEDLINE | ID: mdl-38472643

ABSTRACT

The regenerative function of stem cells is compromised when the proportion of senescent stem cells increases with ageing advance. Therefore, combating stem cell senescence is of great importance for stem cell-based tissue engineering in the elderly, but remains largely unexplored. Osteopontin (OPN), a glycosylated phosphoprotein, is one of the key extracellular matrix molecules in bone tissue. OPN activates various signalling pathways and modulates cellular activities, including cell senescence. However, the role of OPN in stem cell senescence remains largely unknown. This study aims to investigate if OPN modulates cell senescence and bone regenerative function in human adipose-derived mesenchymal stem cells (ASCs), and to determine the underlying mechanisms. We first developed a senescent ASC model using serial passaging until passage 10 (P10), in which senescent cells were characterised by reduced proliferation and osteogenic differentiation capacity compared to P4 ASCs. The conditioned medium from P10 ASCs exhibited a diminished trophic effect on human osteoblasts (HOBs), compared to that from P4 ASCs. P10 ASCs on OPN-coated surface showed rejuvenated phenotype and enhanced osteogenic differentiation. The conditioned medium from P10 ASCs on OPN-coating improved trophic effects on HOBs. OPN regulated the morphology of senescent ASCs, transforming them from a more rounded and flattened cell shape to an elongated shape with a smaller area. These findings demonstrated the effects of OPN in restoring senescent ASCs functions, possibly through a mechanism that involves the modulation of cell morphology, indicating that OPN might hold a great potential for rejuvenating senescent stem cells and could potentially open a new venue for regenerating bone tissue in age-related diseases.


Subject(s)
Adipose Tissue , Bone Regeneration , Cell Differentiation , Cellular Senescence , Mesenchymal Stem Cells , Osteogenesis , Osteopontin , Osteopontin/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Adipose Tissue/cytology , Cell Proliferation/drug effects , Osteoblasts/metabolism , Osteoblasts/cytology , Cells, Cultured , Culture Media, Conditioned/pharmacology , Bone and Bones/cytology , Bone and Bones/metabolism
4.
Int J Biol Macromol ; 266(Pt 1): 131167, 2024 May.
Article in English | MEDLINE | ID: mdl-38547948

ABSTRACT

Blend polymers composed of natural polymers are a ubiquitous biomaterial class due to their suitable mechanical and biological characterization. In the present study, composite scaffolds based on bacterial cellulose (BC)/silk fibroin (SF) with bioactive glass nanoparticles (BGNPs) were developed to enhance osteogenesis in human adipose derived stem cells (hASCs). The scanning electron microscopy (SEM) results of BGNPs indicated a spherical morphology and size ranging from 15 to 30 nm. The presence of BC and BGNPs reduced the pore diameter of SF scaffolds to about 210 ± 10 µm and 205 ± 10 µm, respectively, while increasing their compressive strength and compressive modulus. FTIR analyses proved the presence of BGNPs, BC and SF in the scaffolds. Flow cytometry data confirmed the surface markers for hASCs. The results also showed that BC and BGNPs addition to BC/SF scaffolds decreased degradation and swelling rate. The gene expression (Runx2, alkaline phosphatase and osteocalcin) studies signified the osteogenic potential of BGNPs in BC/SF scaffolds on hASCs. Eventually, the increased cell adhesion, viability and differentiation in the BC/SF and BC/SF/BGNPs composite scaffolds drawn from MTT, SEM, Alizarin red staining and alkaline phosphatase activity confirmed that these scaffolds promise to serve as a therapeutic candidate for bone defects.


Subject(s)
Cellulose , Fibroins , Nanoparticles , Osteogenesis , Tissue Engineering , Tissue Scaffolds , Fibroins/chemistry , Fibroins/pharmacology , Osteogenesis/drug effects , Tissue Scaffolds/chemistry , Cellulose/chemistry , Cellulose/pharmacology , Humans , Tissue Engineering/methods , Nanoparticles/chemistry , Glass/chemistry , Cell Differentiation/drug effects , Bone and Bones/drug effects , Bone and Bones/cytology , Bone and Bones/metabolism , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Stem Cells/drug effects , Stem Cells/cytology , Stem Cells/metabolism , Cell Adhesion/drug effects , Cell Survival/drug effects , Cell Proliferation/drug effects , Alkaline Phosphatase/metabolism
5.
Exp Cell Res ; 431(1): 113751, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37574037

ABSTRACT

Primary cilia are non-motile, microtubule-based sensory organelle present in most vertebrate cells with a fundamental role in the modulation of organismal development, morphogenesis, and repair. Here we focus on the role of primary cilia in embryonic and postnatal skeletal development. We examine evidence supporting its involvement in physiochemical and developmental signaling that regulates proliferation, patterning, differentiation and homeostasis of osteoblasts, chondrocytes, and their progenitor cells in the skeleton. We discuss how signaling effectors in mechanotransduction and bone development, such as Hedgehog, Wnt, Fibroblast growth factor and second messenger pathways operate at least in part at the primary cilium. The relevance of primary cilia in bone formation and maintenance is underscored by a growing list of rare genetic skeletal ciliopathies. We collate these findings and summarize the current understanding of molecular factors and mechanisms governing primary ciliogenesis and ciliary function in skeletal development and disease.


Subject(s)
Bone and Bones , Skeleton , Cilia , Humans , Animals , Bone and Bones/cytology , Bone and Bones/pathology , Skeleton/growth & development , Organogenesis , Osteogenesis , Signal Transduction , Mechanotransduction, Cellular
6.
J Biol Chem ; 299(6): 104805, 2023 06.
Article in English | MEDLINE | ID: mdl-37172728

ABSTRACT

Bone development starts with condensations of undifferentiated mesenchymal cells that set a framework for future bones within the primordium. In the endochondral pathway, mesenchymal cells inside the condensation differentiate into chondrocytes and perichondrial cells in a SOX9-dependent mechanism. However, the identity of mesenchymal cells outside the condensation and how they participate in developing bones remain undefined. Here we show that mesenchymal cells surrounding the condensation contribute to both cartilage and perichondrium, robustly generating chondrocytes, osteoblasts, and marrow stromal cells in developing bones. Single-cell RNA-seq analysis of Prrx1-cre-marked limb bud mesenchymal cells at E11.5 reveals that Notch effector Hes1 is expressed in a mutually exclusive manner with Sox9 that is expressed in pre-cartilaginous condensations. Analysis of a Notch signaling reporter CBF1:H2B-Venus reveals that peri-condensation mesenchymal cells are active for Notch signaling. In vivo lineage-tracing analysis using Hes1-creER identifies that Hes1+ early mesenchymal cells surrounding the SOX9+ condensation at E10.5 contribute to both cartilage and perichondrium at E13.5, subsequently becoming growth plate chondrocytes, osteoblasts of trabecular and cortical bones, and marrow stromal cells in postnatal bones. In contrast, Hes1+ cells in the perichondrium at E12.5 or E14.5 do not generate chondrocytes within cartilage, contributing to osteoblasts and marrow stromal cells only through the perichondrial route. Therefore, Hes1+ peri-condensation mesenchymal cells give rise to cells of the skeletal lineage through cartilage-dependent and independent pathways, supporting the theory that early mesenchymal cells outside the condensation also play important roles in early bone development.


Subject(s)
Bone Development , Bone and Bones , Cartilage , Cell Differentiation , Cell Lineage , Chondrocytes , Mesenchymal Stem Cells , Transcription Factor HES-1 , Animals , Mice , Bone and Bones/cytology , Cartilage/cytology , Cartilage/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Transcription Factor HES-1/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Receptors, Notch/metabolism
7.
Front Endocrinol (Lausanne) ; 14: 1145467, 2023.
Article in English | MEDLINE | ID: mdl-37181042

ABSTRACT

Introduction: The view that bone and energy metabolism are integrated by common regulatory mechanisms is broadly accepted and supported by multiple strands of evidence. This includes the well-characterized role of the PPARγ nuclear receptor, which is a common denominator in energy metabolism and bone metabolism. Little is known, however, about the role of PPARα nuclear receptor, a major regulator of lipid metabolism in other organs, in bone. Methods: A side-by-side comparative study of 5-15 mo old mice with global PPARα deficiency (αKO) and mice with osteocyte-specific PPARα deficiency (αOTKO) in order to parse out the various activities of PPARα in the skeleton that are of local and systemic significance. This study included transcriptome analysis of PPARα-deficient osteocytes, and analyses of bone mass and bone microarchitecture, systemic energy metabolism with indirect calorimetry, and differentiation potential of hematopoietic and mesenchymal bone cell progenitors. These analyses were paired with in vitro studies of either intact or silenced for PPARα MLO-A5 cells to determine PPARα role in osteocyte bioenergetics. Results: In osteocytes, PPARα controls large number of transcripts coding for signaling and secreted proteins which may regulate bone microenvironment and peripheral fat metabolism. In addition, PPARα in osteocytes controls their bioenergetics and mitochondrial response to stress, which constitutes up to 40% of total PPARα contribution to the global energy metabolism. Similarly to αKO mice, the metabolic phenotype of αOTKO mice (both males and females) is age-dependent. In younger mice, osteocyte metabolism contributes positively to global energetics, however, with aging the high-energy phenotype reverts to a low-energy phenotype and obesity develops, suggesting a longitudinal negative effect of impaired lipid metabolism and mitochondrial dysfunction in osteocytes deficient in PPARα. However, bone phenotype was not affected in αOTKO mice except in the form of an increased volume of marrow adipose tissue in males. In contrast, global PPARα deficiency in αKO mice led to enlarged bone diameter with a proportional increase in number of trabeculae and enlarged marrow cavities; it also altered differentiation of hematopoietic and mesenchymal marrow cells toward osteoclast, osteoblast and adipocyte lineages, respectively. Discussion: PPARα role in bone is multileveled and complex. In osteocytes, PPARα controls the bioenergetics of these cells, which significantly contributes to systemic energy metabolism and their endocrine/paracrine function in controlling marrow adiposity and peripheral fat metabolism.


Subject(s)
Bone and Bones , Energy Metabolism , Osteocytes , PPAR alpha , Osteocytes/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Bone and Bones/cytology , Bone and Bones/metabolism , Energy Metabolism/genetics , Animals , Mice , Cells, Cultured , Male , Female , Signal Transduction , Mice, Knockout , Hematopoietic Stem Cells/cytology , Cell Differentiation/genetics , Age Factors , Gene Expression Profiling
8.
Anat Rec (Hoboken) ; 306(7): 1762-1803, 2023 07.
Article in English | MEDLINE | ID: mdl-35860957

ABSTRACT

We name and describe a new iguanodontian dinosaur from the Early Creteceous Kirkwood Formation, Eastern Cape Province, South Africa. This dinosaur is one of only two ornithopod dinosaurs known from the Cretaceous of southern Africa, and is unique in being represented primarily by hatchling to young juvenile individuals as demonstrated by bone histological analysis. All of the juvenile material of this new taxon comes from a single, laterally-restricted bonebed and specimens were primarily recovered as partial to complete single elements, although rare articulated materials and one partial skeleton were found. Sedimentology of the bonebed suggests that this horizon heralds a change in environment upsection to a drier and more seasonal climate. This accumulation of bones is interpreted as seasonal mortality from a nesting site or nesting grounds and may be linked to this environmental shift.


Subject(s)
Dinosaurs , Animals , Bone and Bones/cytology , Dinosaurs/anatomy & histology , Dinosaurs/classification , Dinosaurs/physiology , South Africa , Fossils
9.
Nat Commun ; 13(1): 3960, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35803931

ABSTRACT

Mesenchymal stromal cells (MSCs) differentiation into different lineages is precisely controlled by signaling pathways. Given that protein kinases play a crucial role in signal transduction, here we show that Microtubule Associated Serine/Threonine Kinase Family Member 4 (Mast4) serves as an important mediator of TGF-ß and Wnt signal transduction in regulating chondro-osteogenic differentiation of MSCs. Suppression of Mast4 by TGF-ß1 led to increased Sox9 stability by blocking Mast4-induced Sox9 serine 494 phosphorylation and subsequent proteasomal degradation, ultimately enhancing chondrogenesis of MSCs. On the other hand, Mast4 protein, which stability was enhanced by Wnt-mediated inhibition of GSK-3ß and subsequent Smurf1 recruitment, promoted ß-catenin nuclear localization and Runx2 activity, increasing osteogenesis of MSCs. Consistently, Mast4-/- mice demonstrated excessive cartilage synthesis, while exhibiting osteoporotic phenotype. Interestingly, Mast4 depletion in MSCs facilitated cartilage formation and regeneration in vivo. Altogether, our findings uncover essential roles of Mast4 in determining the fate of MSC development into cartilage or bone.


Subject(s)
Bone and Bones , Cartilage , Mesenchymal Stem Cells , Microtubule-Associated Proteins , Protein Serine-Threonine Kinases , Animals , Female , Mice , Bone and Bones/cytology , Bone and Bones/metabolism , Cartilage/cytology , Cartilage/metabolism , Cell Differentiation/genetics , Chondrogenesis/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Osteogenesis/genetics , Protein Serine-Threonine Kinases/genetics , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway
10.
Signal Transduct Target Ther ; 7(1): 155, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35538062

ABSTRACT

Maxillofacial bone defects are commonly seen in clinical practice. A clearer understanding of the regulatory network directing maxillofacial bone formation will promote the development of novel therapeutic approaches for bone regeneration. The fibroblast growth factor (FGF) signalling pathway is critical for the development of maxillofacial bone. Klotho, a type I transmembrane protein, is an important components of FGF receptor complexes. Recent studies have reported the presence of Klotho expression in bone. However, the role of Klotho in cranioskeletal development and repair remains unknown. Here, we use a genetic strategy to report that deletion of Klotho in Osx-positive mesenchymal progenitors leads to a significant reduction in osteogenesis under physiological and pathological conditions. Klotho-deficient mensenchymal progenitors also suppress osteoclastogenesis in vitro and in vivo. Under conditions of inflammation and trauma-induced bone loss, we find that Klotho exerts an inhibitory function on inflammation-induced TNFR signaling by attenuating Rankl expression. More importantly, we show for the first time that Klotho is present in human alveolar bone, with a distinct expression pattern under both normal and pathological conditions. In summary, our results identify the mechanism whereby Klotho expressed in Osx+-mensenchymal progenitors controls osteoblast differentiation and osteoclastogenesis during mandibular alveolar bone formation and repair. Klotho-mediated signaling is an important component of alveolar bone remodeling and regeneration. It may also be a target for future therapeutics.


Subject(s)
Bone Development , Bone and Bones , Klotho Proteins , Mesenchymal Stem Cells , Osteogenesis , Bone Development/physiology , Bone and Bones/cytology , Bone and Bones/metabolism , Fibroblast Growth Factors/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Klotho Proteins/metabolism , Maxilla/growth & development , Maxilla/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Sp7 Transcription Factor
11.
Calcif Tissue Int ; 110(6): 746-758, 2022 06.
Article in English | MEDLINE | ID: mdl-35137272

ABSTRACT

Ebfs are a family of transcription factors regulating the differentiation of multiple cell types of mesenchymal origin, including osteoblasts. Global deletion of Ebf1 results in increased bone formation and bone mass, while global loss of Ebf2 leads to enhanced bone resorption and decreased bone mass. Targeted deletion of Ebf1 in early committed osteoblasts leads to increased bone formation, whereas deletion in mature osteoblasts has no effect. To study the effects of Ebf2 specifically on long bone development, we created a limb bud mesenchyme targeted Ebf2 knockout mouse model by using paired related homeobox gene 1 (Prrx1) Cre. To investigate the possible interplay between Ebf1 and Ebf2, we deleted both Ebf1 and Ebf2 in the cells expressing Prrx1. Mice with Prrx1-targeted deletion of Ebf2 had a very mild bone phenotype. However, deletion of both Ebf1 and Ebf2 in mesenchymal lineage cells lead to significant, age progressive increase in bone volume. The phenotype was to some extent gender dependent, leading to an increase in both trabecular and cortical bone in females, while in males a mild cortical bone phenotype and a growth plate defect was observed. The phenotype was observed at both 6 and 12 weeks of age, but it was more pronounced in older female mice. Our data suggest that Ebfs modulate bone homeostasis and they are likely able to compensate for the lack of each other. The roles of Ebfs in bone formation appear to be complex and affected by multiple factors, such as age and gender.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Bone and Bones , Homeodomain Proteins , Mesenchymal Stem Cells , Trans-Activators , Age Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone and Bones/cytology , Bone and Bones/metabolism , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Phenotype , Sex Factors , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism
12.
Molecules ; 27(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35163869

ABSTRACT

This prospective clinical study aimed to evaluate the peri-implant hard tissue dimensional change at 6 months of immediate implant placement with bone graft materials in the posterior area using cone-beam computed tomography (CBCT). Twelve dental implants were placed concurrently following tooth extraction in the posterior area and filled with xenograft particles. The CBCT images were taken immediately after surgical procedures and then at 6 months follow-up. To evaluate the hard tissue changes, the vertical and horizontal bone thickness were analyzed and measured using ImageJ software. Paired t-test or Wilcoxon match-pair signed-rank test was done to analyze the changes of hard tissue values at the same level between immediately and 6 months following immediate implant placement. Independent t-test or Mann-Whitney U test was used to analyze the dimensional change in the vertical and horizontal direction in buccal and lingual aspects. The level of significance was set at p value = 0.05. All implants were successfully osseointegrated. At 6 months follow-up, the vertical bone change at the buccal aspect was -0.69 mm and at the lingual aspect -0.39 mm. For horizontal bone thickness, the bone dimensional changes at 0, 1, 5, and 9 mm levels from the implant platform were -0.62 mm, -0.70 mm, -0.24 mm, and -0.22 mm, respectively. A significant bone reduction was observed in all measurement levels during the 6 months after implant placement (p value < 0.05). It was noted that even with bone grafting, a decrease in bone thickness was seen following the immediate implant placement. Therefore, this technique can be an alternative method to place the implant in the posterior area.


Subject(s)
Bone Regeneration , Bone Transplantation/methods , Bone and Bones/cytology , Cone-Beam Computed Tomography/methods , Dental Restoration Repair/methods , Prostheses and Implants/statistics & numerical data , Tooth Attrition/therapy , Adult , Aged , Bone and Bones/diagnostic imaging , Dental Caries/therapy , Humans , Image Processing, Computer-Assisted/methods , Middle Aged , Prospective Studies , Tooth, Nonvital/therapy
13.
Nat Commun ; 13(1): 571, 2022 01 28.
Article in English | MEDLINE | ID: mdl-35091558

ABSTRACT

Developmental osteogenesis, physiological bone remodelling and fracture healing require removal of matrix and cellular debris. Osteoclasts generated by the fusion of circulating monocytes degrade bone, whereas the identity of the cells responsible for cartilage resorption is a long-standing and controversial question. Here we show that matrix degradation and chondrocyte phagocytosis are mediated by fatty acid binding protein 5-expressing cells representing septoclasts, which have a mesenchymal origin and are not derived from haematopoietic cells. The Notch ligand Delta-like 4, provided by endothelial cells, is necessary for septoclast specification and developmental bone growth. Consistent with the termination of growth, septoclasts disappear in adult and ageing bone, but re-emerge in association with growing vessels during fracture healing. We propose that cartilage degradation is mediated by rare, specialized cells distinct from osteoclasts. Our findings have implications for fracture healing, which is frequently impaired in aging humans.


Subject(s)
Cartilage/metabolism , Fracture Healing/physiology , Mesenchymal Stem Cells/metabolism , Osteoclasts/metabolism , Osteogenesis/physiology , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Bone and Bones/ultrastructure , Cartilage/cytology , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Fracture Healing/genetics , Humans , Male , Mesenchymal Stem Cells/cytology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Immunoelectron , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Osteoclasts/cytology , Osteogenesis/genetics , RNA-Seq/methods
14.
Tissue Cell ; 74: 101712, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34920234

ABSTRACT

Fabrication of immunocompatible tissue constructs for bone-cartilage defect regeneration is of prime importance. In this study, a double layer hydrogel was successfully synthesized, where alginate/polyacrylamide were formulated to represent cartilage layer (5-10 % (w/w) total polymer ratio) and laponite XLS (2-5-8% (w/w))/alginate/polyacrylamide formed bone layer. Hydrogels were dried by supercritical CO2 at 100 and 200 bar, 45 °C, 5 g/min CO2 flow rate for 2 h. Constructs were treated with collagen, then cellularized and embedded in cell-laden GelMA to mimic the cellular microenvironment. The optimum weight ratio of alginate/polyacrylamide:laponite XLS was 10:5 based on mechanical strength test results. The constructs yielded high porosity (91.50 m2/g) and mesoporous structure, owing to the diffusivity of CO2 at 200 bar (0.49 × 10-7 m2/s). Constructs were then treated with collagen to increase cell adhesion and ATDC5 cells were seeded in the cartilage layer, whereas hFOB cells to the bone layer. About 10-15 % higher cell viability was attained. The porous structure of the construct allowed infiltration of macrophages, promoted polarization and positively affected the behavior of macrophages, yielding a decrease in M1 markers, whereas an increase in M2 on day 4. The formulated tissue constructs would be of value in tissue engineering applications.


Subject(s)
Acrylic Resins/chemistry , Alginates/chemistry , Bone and Bones/immunology , Carbon Dioxide/chemistry , Hydrogels/chemistry , Macrophages/immunology , Silicates/chemistry , Tissue Engineering , Tissue Scaffolds/chemistry , Animals , Bone and Bones/cytology , Cell Line, Tumor , Humans , Macrophages/cytology , Mice , Porosity , RAW 264.7 Cells
15.
EMBO J ; 41(4): e108415, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34957577

ABSTRACT

Leptin receptor (LepR)-positive cells are key components of the bone marrow hematopoietic microenvironment, and highly enrich skeletal stem and progenitor cells that maintain homeostasis of the adult skeleton. However, the heterogeneity and lineage hierarchy within this population has been elusive. Using genetic lineage tracing and single-cell RNA sequencing, we found that Lepr-Cre labels most bone marrow stromal cells and osteogenic lineage cells in adult long bones. Integrated analysis of Lepr-Cre-traced cells under homeostatic and stress conditions revealed dynamic changes of the adipogenic, osteogenic, and periosteal lineages. Importantly, we discovered a Notch3+ bone marrow sub-population that is slow-cycling and closely associated with the vasculatures, as well as key transcriptional networks promoting osteo-chondrogenic differentiation. We also identified a Sca-1+ periosteal sub-population with high clonogenic activity but limited osteo-chondrogenic potential. Together, we mapped the transcriptomic landscape of adult LepR+ stem and progenitor cells and uncovered cellular and molecular mechanisms underlying their maintenance and lineage specification.


Subject(s)
Bone and Bones/cytology , Receptors, Leptin/metabolism , Single-Cell Analysis/methods , Stem Cells/physiology , Aging/physiology , Animals , Antigens, Ly/metabolism , Cell Differentiation , Cell Lineage , Colony-Forming Units Assay , Female , Fractures, Bone , Gene Expression Profiling , Homeodomain Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Rosiglitazone/pharmacology , Stem Cells/cytology , Stem Cells/drug effects , Stress, Physiological
16.
Exp Cell Res ; 410(2): 112969, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34883111

ABSTRACT

MEL1 (MDS1/EVI1-like gene 1/PRDM16), a zinc finger protein, is located near the chromosomal breakpoint at 1p36 in human acute myeloid leukemia (AML) cells with the t (1; 3) (p36; q21) translocation. Mel1/Prdm16 is not only a causative gene of leukemia, but also has multiple regulatory functions, such as the regulation of fat metabolism. To investigate the function of Mel1/Prdm16, we generated Mel1/Prdm16-deficient mice, but homozygous deficiency (Mel1/Prdm16-/-) was embryonic lethal at E 11.5. Heterozygous mice showed abnormal cartilage and bone formation in the postnatal skull and long bones, suggesting that Mel1/Prdm16 expression plays an important role in bone development. In osteoblast and chondrocyte cell lines, Mel1/Prdm16 promotes the differentiation of chondrocytes and regulates the differentiation of osteoblasts. Transient repression of the master regulator Runx2 is required for chondrocyte differentiation at an early stage of differentiation. However, in Mel1/Prdm16-suppressed ATDC5 cells, the initial suppression of Runx2 was lacking and its expression was upregulated at the beginning of differentiation, suggesting that chondrogenic differentiation is suppressed in Mel1/Prdm16+/- mesenchymal progenitor cells because Runx2 expression is upregulated during the early stage of differentiation. Thus, the Mel1/Prdm16 gene may be involved in the early repression of Runx2 expression during osteochondral differentiation and promote chondrogenic differentiation.


Subject(s)
Bone and Bones/anatomy & histology , Bone and Bones/cytology , Cell Differentiation , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Bone Morphogenetic Protein 2/metabolism , Cartilage/pathology , Core Binding Factor Alpha 1 Subunit/metabolism , DNA-Binding Proteins/deficiency , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Models, Biological , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Signal Transduction , Transcription Factors/deficiency
17.
Aging (Albany NY) ; 13(24): 25607-25642, 2021 12 30.
Article in English | MEDLINE | ID: mdl-34968192

ABSTRACT

Aging is accompanied by osteopenia, characterized by reduced bone formation and increased bone resorption. Osteocytes, the terminally differentiated osteoblasts, are regulators of bone homeostasis, and parathyroid hormone (PTH) receptor (PPR) signaling in mature osteoblasts/osteocytes is essential for PTH-driven anabolic and catabolic skeletal responses. However, the role of PPR signaling in those cells during aging has not been investigated. The aim of this study was to analyze the role of PTH signaling in mature osteoblasts/osteocytes during aging. Mice lacking PPR in osteocyte (Dmp1-PPRKO) display an age-dependent osteopenia characterized by a significant decrease in osteoblast activity and increase in osteoclast number and activity. At the molecular level, the absence of PPR signaling in mature osteoblasts/osteocytes is associated with an increase in serum sclerostin and a significant increase in osteocytes expressing 4-hydroxy-2-nonenals, a marker of oxidative stress. In Dmp1-PPRKO mice there was an age-dependent increase in p16Ink4a/Cdkn2a expression, whereas it was unchanged in controls. In vitro studies demonstrated that PTH protects osteocytes from oxidative stress-induced cell death. In summary, we reported that PPR signaling in osteocytes is important for protecting the skeleton from age-induced bone loss by restraining osteoclast's activity and protecting osteocytes from oxidative stresses.


Subject(s)
Osteoblasts/drug effects , Osteoclasts/drug effects , Osteocytes/drug effects , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction/drug effects , Animals , Bone Diseases, Metabolic/pathology , Bone Resorption/metabolism , Bone and Bones/cytology , Bone and Bones/drug effects , Bone and Bones/metabolism , Homeostasis/drug effects , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Osteocytes/metabolism , Osteoporosis/metabolism
18.
J Mater Sci Mater Med ; 32(12): 148, 2021 Dec 04.
Article in English | MEDLINE | ID: mdl-34862915

ABSTRACT

There is continued focus on the development of new biomaterials and associated biological testing methods needed to reduce the time taken for their entry to clinical use. The application of Raman spectroscopy to the study of individual cells that have been in contact with biomaterials offers enhanced in vitro information in a potentially non-destructive testing regime. The work presented here reports the Raman spectral analysis of discreet U-2 OS bone cells after exposure to hydroxyapatite (HA) coated titanium (Ti) substrates in both the as-deposited and thermally annealed states. These data show that cells that were in contact with the bioactive HA surface for 7 days had spectral markers similar to those cultured on the Ti substrate control for the same period. However, the spectral features for those cells that were in contact with the annealed HA surface had indicators of significant differentiation at day 21 while cells on the as-deposited surface did not show these Raman changes until day 28. The cells adhered to pristine Ti control surface showed no spectral changes at any of the timepoints studied. The validity of these spectroscopic results has been confirmed using data from standard in vitro cell viability, adhesion, and proliferation assays over the same 28-day culture period. In this case, cell maturation was evidenced by the formation of natural bone apatite, which precipitated intracellularly for cells exposed to both types of HA-coated Ti at 21 and 28 days, respectively. The properties of the intracellular apatite were markedly different from that of the synthetic HA used to coat the Ti substrate with an average particle size of 230 nm, a crystalline-like shape and Ca/P ratio of 1.63 ± 0.5 as determined by SEM-EDX analysis. By comparison, the synthetic HA particles used as a control had an average size of 372 nm and were more-rounded in shape with a Ca/P ratio of 0.8 by XPS analysis and 1.28 by SEM-EDX analysis. This study shows that Raman spectroscopy can be employed to monitor single U-2 OS cell response to biomaterials that promote cell maturation towards de novo bone thereby offering a label-free in vitro testing method that allows for non-destructive analyses.


Subject(s)
Bone and Bones/cytology , Durapatite/pharmacology , Single-Cell Analysis , Spectrum Analysis, Raman , Titanium/pharmacology , Biocompatible Materials , Biomarkers , Cell Line , Humans , Materials Testing
19.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34884519

ABSTRACT

Magnesium (Mg2+) is known to play a crucial role in mineral and matrix metabolism of bone tissue and is thus increasingly considered in the field of bone tissue engineering. Bioactive glasses (BGs) offer the promising possibility of the incorporation and local delivery of therapeutically active ions as Mg2+. In this study, two Mg2+-doped derivatives of the ICIE16-BG composition (49.46 SiO2, 36.27 CaO, 6.6 Na2O, 1.07 P2O5, 6.6 K2O (mol%)), namely 6Mg-BG (49.46 SiO2, 30.27 CaO, 6.6 Na2O, 1.07 P2O5, 6.6 K2O, 6.0 MgO (mol%) and 3Mg-BG (49.46 SiO2, 33.27 CaO, 6.6 Na2O, 1.07 P2O5, 6.6 K2O, 3.0 MgO (mol%)) were examined. Their influence on viability, proliferation and osteogenic differentiation of human mesenchymal stromal cells (MSCs) was explored in comparison to the original ICIE16-BG. All BGs showed good biocompatibility. The Mg2+-doped BGs had a positive influence on MSC viability alongside with inhibiting effects on MSC proliferation. A strong induction of osteogenic differentiation markers was observed, with the Mg2+-doped BGs significantly outperforming the ICIE16-BG regarding the expression of genes encoding for protein members of the osseous extracellular matrix (ECM) at certain observation time points. However, an overall Mg2+-induced enhancement of the expression of genes encoding for ECM proteins could not be observed, possibly due to a too moderate Mg2+ release. By adaption of the Mg2+ release from BGs, an even stronger impact on the expression of genes encoding for ECM proteins might be achieved. Furthermore, other BG-types such as mesoporous BGs might provide a higher local presence of the therapeutically active ions and should therefore be considered for upcoming studies.


Subject(s)
Bone and Bones/cytology , Cell Differentiation , Glass/chemistry , Magnesium/chemistry , Mesenchymal Stem Cells/cytology , Osteogenesis , Tissue Engineering/methods , Cell Proliferation , Humans , In Vitro Techniques
20.
Int J Mol Sci ; 22(23)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34884557

ABSTRACT

Octacalcium phosphate (OCP, Ca8H2(PO4)6·5H2O) is known to be a possible precursor of biological hydroxyapatite formation of organic bone tissue. OCP has higher biocompatibility and osseointegration rate compared to other calcium phosphates. In this work, the synthesis of low-temperature calcium phosphate compounds and substituted forms of those at physiological temperatures is shown. Strontium is used to improve bioactive properties of the material. Strontium was inserted into the OCP structure by ionic substitution in solutions. The processes of phase formation of low-temperature OCP with theoretical substitution of strontium for calcium up to 50 at.% in conditions close to physiological, i.e., temperature 35-37 °C and normal pressure, were described. The effect of strontium substitution range on changes in the crystal lattice of materials, the microstructural features, surface morphology and biological properties in vitro has been established. The results of the study indicate the effectiveness of using strontium in OCP for improving biocompatibility of OCP based composite materials intended for bone repair.


Subject(s)
Biocompatible Materials/pharmacology , Bone Regeneration , Bone and Bones/cytology , Calcium Phosphates/chemical synthesis , Calcium Phosphates/pharmacology , Mesoderm/cytology , Animals , Biocompatible Materials/chemical synthesis , Bone and Bones/drug effects , Durapatite/chemistry , In Vitro Techniques , Mesoderm/drug effects , Mice , Mice, Inbred C3H , Reactive Oxygen Species/metabolism , Strontium/chemistry , Tissue Engineering
SELECTION OF CITATIONS
SEARCH DETAIL
...