Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.256
Filter
1.
Development ; 151(7)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38602508

ABSTRACT

The skull roof, or calvaria, is comprised of interlocking plates of bones that encase the brain. Separating these bones are fibrous sutures that permit growth. Currently, we do not understand the instructions for directional growth of the calvaria, a process which is error-prone and can lead to skeletal deficiencies or premature suture fusion (craniosynostosis, CS). Here, we identify graded expression of fibronectin (FN1) in the mouse embryonic cranial mesenchyme (CM) that precedes the apical expansion of calvaria. Conditional deletion of Fn1 or Wasl leads to diminished frontal bone expansion by altering cell shape and focal actin enrichment, respectively, suggesting defective migration of calvarial progenitors. Interestingly, Fn1 mutants have premature fusion of coronal sutures. Consistently, syndromic forms of CS in humans exhibit dysregulated FN1 expression, and we also find FN1 expression altered in a mouse CS model of Apert syndrome. These data support a model of FN1 as a directional substrate for calvarial osteoblast migration that may be a common mechanism underlying many cranial disorders of disparate genetic etiologies.


Subject(s)
Fibronectins , Premature Birth , Skull , Animals , Female , Humans , Mice , Cues , Disease Models, Animal , Fibronectins/metabolism , Osteoblasts , Skull/cytology , Skull/growth & development , Skull/metabolism , Sutures
2.
Cell ; 186(17): 3706-3725.e29, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37562402

ABSTRACT

The bone marrow in the skull is important for shaping immune responses in the brain and meninges, but its molecular makeup among bones and relevance in human diseases remain unclear. Here, we show that the mouse skull has the most distinct transcriptomic profile compared with other bones in states of health and injury, characterized by a late-stage neutrophil phenotype. In humans, proteome analysis reveals that the skull marrow is the most distinct, with differentially expressed neutrophil-related pathways and a unique synaptic protein signature. 3D imaging demonstrates the structural and cellular details of human skull-meninges connections (SMCs) compared with veins. Last, using translocator protein positron emission tomography (TSPO-PET) imaging, we show that the skull bone marrow reflects inflammatory brain responses with a disease-specific spatial distribution in patients with various neurological disorders. The unique molecular profile and anatomical and functional connections of the skull show its potential as a site for diagnosing, monitoring, and treating brain diseases.


Subject(s)
Bone Marrow , Nervous System Diseases , Skull , Animals , Humans , Mice , Bone Marrow/metabolism , Brain/diagnostic imaging , Brain/metabolism , Carrier Proteins/metabolism , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Positron-Emission Tomography/methods , Receptors, GABA/metabolism , Skull/cytology , Skull/diagnostic imaging
3.
BMC Mol Cell Biol ; 24(1): 9, 2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36890454

ABSTRACT

BACKGROUND: Mechanical therapies, such as distraction osteogenesis, are widely used in dental clinics. During this process, the mechanisms by which tensile force triggers bone formation remain of interest. Herein, we investigated the influence of cyclic tensile stress on osteoblasts and identified the involvement of ERK1/2 and STAT3. MATERIALS AND METHODS: Rat clavarial osteoblasts were subjected to tensile loading (10% elongation, 0.5 Hz) for different time periods. RNA and protein levels of osteogenic markers were determined using qPCR and western blot after inhibition of ERK1/2 and STAT3. ALP activity and ARS staining revealed osteoblast mineralization capacity. The interaction between ERK1/2 and STAT3 was investigated by immunofluorescence, western blot, and Co-IP. RESULTS: The results showed that tensile loading significantly promoted osteogenesis-related genes, proteins and mineralized nodules. In loading-induced osteoblasts, inhibition of ERK1/2 or STAT3 decreased osteogenesis-related biomarkers significantly. Moreover, ERK1/2 inhibition suppressed STAT3 phosphorylation, and STAT3 inhibition disrupted the nuclear translocation of pERK1/2 induced by tensile loading. In the non-loading environment, inhibition of ERK1/2 hindered osteoblast differentiation and mineralization, while STAT3 phosphorylation was elevated after ERK1/2 inhibition. STAT3 inhibition also increased ERK1/2 phosphorylation, but did not significantly affect osteogenesis-related factors. CONCLUSION: Taken together, these data suggested that ERK1/2 and STAT3 interacted in osteoblasts. ERK1/2-STAT3 were sequentially activated by tensile force loading, and both affected osteogenesis during the process.


Subject(s)
Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Osteoblasts , STAT3 Transcription Factor , Skull , Animals , Rats , Cells, Cultured , MAP Kinase Signaling System , Osteoblasts/metabolism , Osteogenesis , Phosphorylation , STAT3 Transcription Factor/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Skull/cytology , Skull/metabolism
4.
Development ; 149(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35132438

ABSTRACT

Cranial neural crest cell (NCC)-derived chondrocyte precursors undergo a dynamic differentiation and maturation process to establish a scaffold for subsequent bone formation, alterations in which contribute to congenital birth defects. Here, we demonstrate that transcription factor and histone methyltransferase proteins Prdm3 and Prdm16 control the differentiation switch of cranial NCCs to craniofacial cartilage. Loss of either paralog results in hypoplastic and disorganized chondrocytes due to impaired cellular orientation and polarity. We show that these proteins regulate cartilage differentiation by controlling the timing of Wnt/ß-catenin activity in strikingly different ways: Prdm3 represses whereas Prdm16 activates global gene expression, although both act by regulating Wnt enhanceosome activity and chromatin accessibility. Finally, we show that manipulating Wnt/ß-catenin signaling pharmacologically or generating prdm3-/-;prdm16-/- double mutants rescues craniofacial cartilage defects. Our findings reveal upstream regulatory roles for Prdm3 and Prdm16 in cranial NCCs to control Wnt/ß-catenin transcriptional activity during chondrocyte differentiation to ensure proper development of the craniofacial skeleton.


Subject(s)
Cell Differentiation , MDS1 and EVI1 Complex Locus Protein/metabolism , Wnt Signaling Pathway/genetics , Zebrafish Proteins/metabolism , Animals , Cartilage/cytology , Cartilage/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , MDS1 and EVI1 Complex Locus Protein/deficiency , MDS1 and EVI1 Complex Locus Protein/genetics , Mice , Mice, Knockout , Neural Crest/cytology , Neural Crest/metabolism , Regulatory Sequences, Nucleic Acid , Skull/cytology , Skull/metabolism , Wnt Proteins/metabolism , Zebrafish , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , beta Catenin/metabolism
5.
Nat Commun ; 13(1): 13, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013168

ABSTRACT

The cranial neural crest generates a huge diversity of derivatives, including the bulk of connective and skeletal tissues of the vertebrate head. How neural crest cells acquire such extraordinary lineage potential remains unresolved. By integrating single-cell transcriptome and chromatin accessibility profiles of cranial neural crest-derived cells across the zebrafish lifetime, we observe progressive and region-specific establishment of enhancer accessibility for distinct fates. Neural crest-derived cells rapidly diversify into specialized progenitors, including multipotent skeletal progenitors, stromal cells with a regenerative signature, fibroblasts with a unique metabolic signature linked to skeletal integrity, and gill-specific progenitors generating cell types for respiration. By retrogradely mapping the emergence of lineage-specific chromatin accessibility, we identify a wealth of candidate lineage-priming factors, including a Gata3 regulatory circuit for respiratory cell fates. Rather than multilineage potential being established during cranial neural crest specification, our findings support progressive and region-specific chromatin remodeling underlying acquisition of diverse potential.


Subject(s)
Cell Differentiation/physiology , Neural Crest , Single-Cell Analysis , Zebrafish/embryology , Animals , Chromatin , Gene Expression Regulation, Developmental , Neural Crest/cytology , Neural Crest/metabolism , Single-Cell Analysis/methods , Skull/cytology , Transcriptome , Zebrafish/metabolism
6.
Sci Rep ; 11(1): 21907, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34754046

ABSTRACT

Cell-based therapy using mesenchymal stem cells (MSCs) is a novel treatment strategy for spinal cord injury (SCI). MSCs can be isolated from various tissues, and their characteristics vary based on the source. However, reports demonstrating the effect of transplanted rat cranial bone-derived MSCs (rcMSCs) on rat SCI models are lacking. In this study, we determined the effect of transplanting rcMSCs in rat SCI models. MSCs were established from collected bone marrow and cranial bones. SCI rats were established using the weight-drop method and transplanted intravenously with MSCs at 24 h post SCI. The recovery of motor function and hindlimb electrophysiology was evaluated 4 weeks post transplantation. Electrophysiological recovery was evaluated by recording the transcranial electrical stimulation motor-evoked potentials. Tissue repair after SCI was assessed by calculating the cavity ratio. The expression of genes involved in the inflammatory response and cell death in the spinal cord tissue was assessed by real-time polymerase chain reaction. The transplantation of rcMSCs improved motor function and electrophysiology recovery, and reduced cavity ratio. The expression of proinflammatory cytokines was suppressed in the spinal cord tissues of the rats that received rcMSCs. These results demonstrate the efficacy of rcMSCs as cell-based therapy for SCI.


Subject(s)
Mesenchymal Stem Cell Transplantation , Skull/cytology , Spinal Cord Injuries/therapy , Animals , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Rats , Recovery of Function , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology
7.
Int J Mol Sci ; 22(21)2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34768759

ABSTRACT

Concentrated growth factor (CGF) is 100% blood-derived, cross-linked fibrin glue with platelets and growth factors. Human CGF clot is transformed into membrane by a compression device, which has been widely used clinically. However, the mechanical properties of the CGF membranes have not been well characterized. The aims of this study were to measure the tensile strength of human CGF membrane and observe its behavior as a scaffold of BMP-2 in ectopic site over the skull. The tensile test of the full length was performed at the speed of 2mm/min. The CGF membrane (5 × 5 × 2 mm3) or the CGF/BMP-2 (1.0 µg) membrane was grafted onto the skull periosteum of nude mice (5-week-old, male), and harvested at 14 days after the graft. The appearance and size of the CGF membranes were almost same for 7 days by soaking at 4 °C in saline. The average values of the tensile strength at 0 day and 7 days were 0.24 MPa and 0.26 MPa, respectively. No significant differences of both the tensile strength and the elastic modulus were found among 0, 1, 3, and 7 days. Supra-periosteal bone induction was found at 14 days in the CGF/BMP-2, while the CGF alone did not induce bone. These results demonstrated that human CGF membrane could become a short-term, sticky fibrin scaffold for BMP-2, and might be preserved as auto-membranes for wound protection after the surgery.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/therapeutic use , Periosteum/drug effects , Skull/drug effects , Adult , Animals , Bone Morphogenetic Protein 2/therapeutic use , Bone Transplantation , Elastic Modulus , Fibrin Tissue Adhesive/chemistry , Fibrin Tissue Adhesive/pharmacology , Fibrin Tissue Adhesive/therapeutic use , Healthy Volunteers , Humans , Intercellular Signaling Peptides and Proteins/isolation & purification , Male , Membranes/chemistry , Membranes/metabolism , Mice, Nude , Periosteum/cytology , Skull/cytology , Tensile Strength , Wound Healing/drug effects
8.
Nat Rev Neurosci ; 22(10): 616-626, 2021 10.
Article in English | MEDLINE | ID: mdl-34471282

ABSTRACT

In their seminal 1983 paper, Gans and Northcutt proposed that evolution of the vertebrate 'new head' was made possible by the advent of the neural crest and cranial placodes. The neural crest is a stem cell population that arises adjacent to the forming CNS and contributes to important cell types, including components of the peripheral nervous system and craniofacial skeleton and elements of the cardiovascular system. In the past few years, the new head hypothesis has been challenged by the discovery in invertebrate chordates of cells with some, but not all, characteristics of vertebrate neural crest cells. Here, we discuss recent findings regarding how neural crest cells may have evolved during the course of deuterostome evolution. The results suggest that there was progressive addition of cell types to the repertoire of neural crest derivatives throughout vertebrate evolution. Novel genomic tools have enabled higher resolution insight into neural crest evolution, from both a cellular and a gene regulatory perspective. Together, these data provide clues regarding the ancestral neural crest state and how the neural crest continues to evolve to contribute to the success of vertebrates as efficient predators.


Subject(s)
Biological Evolution , Gene Expression Regulation, Developmental/physiology , Neural Crest/growth & development , Skull/growth & development , Animals , Humans , Neural Crest/cytology , Skull/cytology , Vertebrates
9.
Neurosci Lett ; 763: 136179, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34416344

ABSTRACT

Neural differentiation of mesenchymal stromal cells has been widely studied. However, a comparative characterization of ultrastructural changes during neural differentiation has not been performed. In this study, we conducted scanning electron microscopy and transmission electron microscopy analysis to show the morphological changes in mesenchymal stromal cells upon induction of neural differentiation. In addition, transmission electron microscopy results demonstrated ultrastructural differences between human cranial bone marrow mesenchymal stromal cells and iliac crest bone marrow mesenchymal stromal cells. We propose that enriched microvesicles in cranial bone marrow mesenchymal stromal cells may be responsible for the increased efficiency of neural differentiation.


Subject(s)
Mesenchymal Stem Cells/ultrastructure , Neurogenesis , Skull/cytology , Bone Marrow/blood supply , Cells, Cultured , Humans , Ilium/cytology , Mesenchymal Stem Cells/physiology , Microscopy, Electron, Transmission , Microvessels/anatomy & histology , Primary Cell Culture
10.
Nat Commun ; 12(1): 4797, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34376651

ABSTRACT

Sutures separate the flat bones of the skull and enable coordinated growth of the brain and overlying cranium. The coronal suture is most commonly fused in monogenic craniosynostosis, yet the unique aspects of its development remain incompletely understood. To uncover the cellular diversity within the murine embryonic coronal suture, we generated single-cell transcriptomes and performed extensive expression validation. We find distinct pre-osteoblast signatures between the bone fronts and periosteum, a ligament-like population above the suture that persists into adulthood, and a chondrogenic-like population in the dura mater underlying the suture. Lineage tracing reveals an embryonic Six2+ osteoprogenitor population that contributes to the postnatal suture mesenchyme, with these progenitors being preferentially affected in a Twist1+/-; Tcf12+/- mouse model of Saethre-Chotzen Syndrome. This single-cell atlas provides a resource for understanding the development of the coronal suture and the mechanisms for its loss in craniosynostosis.


Subject(s)
Cranial Sutures/metabolism , Gene Expression Regulation, Developmental , Osteogenesis/genetics , Single-Cell Analysis/methods , Transcriptome/genetics , Acrocephalosyndactylia/embryology , Acrocephalosyndactylia/genetics , Acrocephalosyndactylia/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cranial Sutures/cytology , Cranial Sutures/embryology , Dura Mater/cytology , Dura Mater/embryology , Dura Mater/metabolism , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mice, Knockout , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/metabolism , RNA-Seq/methods , Skull/cytology , Skull/embryology , Skull/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
11.
Int J Mol Sci ; 22(14)2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34299204

ABSTRACT

BACKGROUND: bone tissue regeneration remains a current challenge. A growing body of evidence shows that mitochondrial dysfunction impairs osteogenesis and that this organelle may be the target for new therapeutic options. Current literature illustrates that red and near-infrared light can affect the key cellular pathways of all life forms through interactions with photoacceptors within the cells' mitochondria. The current study aims to provide an understanding of the mechanisms by which photobiomodulation (PBM) by 900-nm wavelengths can induce in vitro molecular changes in pre-osteoblasts. METHODS: The PubMed, Scopus, Cochrane, and Scholar databases were used. The manuscripts included in the narrative review were selected according to inclusion and exclusion criteria. The new experimental set-up was based on irradiation with a 980-nm laser and a hand-piece with a standard Gaussian and flat-top beam profile. MC3T3-E1 pre-osteoblasts were irradiated at 0.75, 0.45, and 0.20 W in continuous-wave emission mode for 60 s (spot-size 1 cm2) and allowed to generate a power density of 0.75, 0.45, and 0.20 W/cm2 and a fluence of 45, 27, and 12 J/cm2, respectively. The frequency of irradiation was once, three times (alternate days), or five times (every day) per week for two consecutive weeks. Differentiation, proliferation, and cell viability and their markers were investigated by immunoblotting, immunolabelling, fluorescein-FragELTM-DNA, Hoechst staining, and metabolic activity assays. RESULTS AND CONCLUSIONS: The 980-nm wavelength can photobiomodulate the pre-osteoblasts, regulating their metabolic schedule. The cellular signal activated by 45 J/cm2, 0.75 W and 0.75 W/cm2 consist of the PI3K/Akt/Bcl-2 pathway; differentiation markers were not affected, nor do other parameters seem to stimulate the cells. Our previous and present data consistently support the window effect of 980 nm, which has also been described in extracted mitochondria, through activation of signalling PI3K/Akt/Bcl-2 and cyclin family, while the Wnt and Smads 2/3-ß-catenin pathway was induced by 55 J/cm2, 0.9 W and 0.9 W/cm2.


Subject(s)
Osteoblasts/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Skull/cytology , Animals , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Lasers , Low-Level Light Therapy/methods , Mice , Osteoblasts/metabolism , Osteoblasts/radiation effects , Osteogenesis , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction , Skull/metabolism , Skull/radiation effects
12.
Methods Mol Biol ; 2308: 177-202, 2021.
Article in English | MEDLINE | ID: mdl-34057724

ABSTRACT

The complex bone marrow microenvironment or niche is an important anatomical structure responsible for hematopoiesis and providing support to the immune cells function. Being the source of immune and blood cells, the interaction of these hematopoietic stem and progenitor cells with the cellular niches regulates their ability for self-renewal, proliferation, and differentiation. Dynamic imaging not only provides spatiotemporal information of cell motility but also the morphological changes due to cell-cell interactions in the bone marrow, providing insights into the ongoing physiological activities within the tissue. Here, we describe customized stages with compatible equipment best suited for the upright two-photon microscope, accompanied by detailed methods for both calvarial and tibial intravital imaging. We demonstrate a general protocol for calvarial imaging using a minimally invasive surgical approach, and introduce a bone shaving-based tibial imaging as a complementary method. To demonstrate the applicability of our method we used Lyz2-EGFP transgenic mice to track bone marrow neutrophil activities as an example.


Subject(s)
Bone Marrow/physiology , Cell Tracking , Hematopoietic Stem Cells/physiology , Intravital Microscopy , Microscopy, Fluorescence, Multiphoton , Neutrophils/physiology , Skull/physiology , Stem Cell Niche , Tibia/physiology , Animals , Bone Marrow/metabolism , Cell Movement , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muramidase/genetics , Muramidase/metabolism , Neutrophils/metabolism , Skull/cytology , Skull/metabolism , Tibia/cytology , Tibia/metabolism
13.
Methods Mol Biol ; 2308: 203-222, 2021.
Article in English | MEDLINE | ID: mdl-34057725

ABSTRACT

Haematopoietic stem cells (HSCs) are instrumental in driving the generation of mature blood cells, essential for various functions including immune defense and tissue remodeling. They reside within a specialised bone marrow (BM) microenvironment , or niche, composed of cellular and chemical components that play key roles in regulating long-term HSC function and survival. While flow cytometry methods have significantly advanced studies of hematopoietic cells, enabling their quantification in steady-state and perturbed situations, we are still learning about the specific BM microenvironments that support distinct lineages and how their niches are altered under stress and with age. Major advances in imaging technology over the last decade have permitted in-depth studies of HSC niches in mice. Here, we describe our protocol for visualizing and analyzing the localization, morphology, and function of niche components in the mouse calvarium, using combined confocal and two-photon intravital microscopy, and we present the specific example of measuring vascular permeability.


Subject(s)
Bone Marrow/physiology , Hematopoietic Stem Cells/physiology , Intravital Microscopy , Microscopy, Confocal , Microscopy, Fluorescence, Multiphoton , Skull/physiology , Stem Cell Niche , Time-Lapse Imaging , Animals , Bone Marrow/metabolism , Capillary Permeability , Genes, Reporter , Hematopoietic Stem Cells/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, Transgenic , Skull/cytology , Skull/metabolism
14.
PLoS Genet ; 17(5): e1009579, 2021 05.
Article in English | MEDLINE | ID: mdl-34033651

ABSTRACT

We sought to understand how perturbation of signaling pathways and their targets generates variable phenotypes. In humans, GATA3 associates with highly variable defects, such as HDR syndrome, microsomia and choanal atresia. We previously characterized a zebrafish point mutation in gata3 with highly variable craniofacial defects to the posterior palate. This variability could be due to residual Gata3 function, however, we observe the same phenotypic variability in gata3 null mutants. Using hsp:GATA3-GFP transgenics, we demonstrate that Gata3 function is required between 24 and 30 hpf. At this time maxillary neural crest cells fated to generate the palate express gata3. Transplantation experiments show that neural crest cells require Gata3 function for palatal development. Via a candidate approach, we determined if Bmp signaling was upstream of gata3 and if this pathway explained the mutant's phenotypic variation. Using BRE:d2EGFP transgenics, we demonstrate that maxillary neural crest cells are Bmp responsive by 24 hpf. We find that gata3 expression in maxillary neural crest requires Bmp signaling and that blocking Bmp signaling, in hsp:DN-Bmpr1a-GFP embryos, can phenocopy gata3 mutants. Palatal defects are rescued in hsp:DN-Bmpr1a-GFP;hsp:GATA3-GFP double transgenic embryos, collectively demonstrating that gata3 is downstream of Bmp signaling. However, Bmp attenuation does not alter phenotypic variability in gata3 loss-of-function embryos, implicating a different pathway. Due to phenotypes observed in hypomorphic shha mutants, the Sonic Hedgehog (Shh) pathway was a promising candidate for this pathway. Small molecule activators and inhibitors of the Shh pathway lessen and exacerbate, respectively, the phenotypic severity of gata3 mutants. Importantly, inhibition of Shh can cause gata3 haploinsufficiency, as observed in humans. We find that gata3 mutants in a less expressive genetic background have a compensatory upregulation of Shh signaling. These results demonstrate that the level of Shh signaling can modulate the phenotypes observed in gata3 mutants.


Subject(s)
Bone Morphogenetic Proteins/genetics , GATA3 Transcription Factor/genetics , Hedgehog Proteins/metabolism , Phenotype , Signal Transduction , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , GATA3 Transcription Factor/metabolism , Haploinsufficiency , Loss of Function Mutation , Mutation , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/metabolism , Organogenesis , Skull/cytology , Skull/embryology , Zebrafish/embryology
15.
Sci Rep ; 11(1): 8502, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33875686

ABSTRACT

Bone is a highly dynamic organ that undergoes remodeling equally regulated by osteoblast-mediated bone formation and osteoclast-mediated bone resorption. To clarify the regulation of osteoblastogenesis, primary murine osteoblasts are required for an in vitro study. Primary osteoblasts are isolated from neonatal calvariae through digestion with collagenase. However, the number of cells collected from one pup is not sufficient for further in vitro experiments, leading to an increase in the use of euthanized pups. We hypothesized that the viscosity of digested calvariae and digestion solution supplemented with collagenase results in cell clumping and reduction of isolated cells from bones. We simply added Benzonase, a genetically engineered endonuclease that shears all forms of DNAs/RNAs, in order to reduce nucleic acid-mediated viscosity. We found that addition of Benzonase increased the number of collected osteoblasts by three fold compared to that without Benzonase through reduction of viscosity. Additionally, Benzonase has no effect on cellular identity and function. The new osteoblast isolation protocol with Benzonase minimizes the number of neonatal pups required for an in vitro study and expands the concept that isolation of other populations of cells including osteocytes that are difficult to be purified could be modified by Benzonase.


Subject(s)
Cell Differentiation , Cell Proliferation , Endonucleases/metabolism , Osteoblasts/cytology , Osteogenesis , Skull/cytology , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Skull/metabolism
16.
Cell Biol Int ; 45(8): 1685-1697, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33811714

ABSTRACT

Electromagnetic fields (EMFs) have emerged as a versatile means for osteoporosis treatment and prevention. However, its optimal application parameters are still elusive. Here, we optimized the frequency parameter first by cell culture screening and then by animal experiment validation. Osteoblasts isolated from newborn rats (ROBs) were exposed 90 min/day to 1.8 mT SEMFs at different frequencies (ranging from 10 to 100 Hz, interval of 10 Hz). SEMFs of 1.8 mT inhibited ROB proliferation at 30, 40, 50, 60 Hz, but increased proliferation at 10, 70, 80 Hz. SEMFs of 10, 50, and 70 Hz promoted ROB osteogenic differentiation and mineralization as shown by alkaline phosphatase (ALP) activity, calcium content, and osteogenesis-related molecule expression analyses, with 50 Hz showing greater effects than 10 and 70 Hz. Treatment of young rats with 1.8 mT SEMFs at 10, 50, or 100 Hz for 2 months significantly increased whole-body bone mineral density (BMD) and femur microarchitecture, with the 50 Hz group showing the greatest effect. Furthermore, 1.8 mT SEMFs extended primary cilia lengths of ROBs and increased protein kinase A (PKA) activation also in a frequency-dependent manner, again with 50 Hz SEMFs showing the greatest effect. Pretreatment of ROBs with the PKA inhibitor KT5720 abolished the effects of SEMFs to increase primary cilia length and promote osteogenic differentiation/mineralization. These results indicate that 1.8 mT SEMFs have a frequency window effect in promoting osteogenic differentiation/mineralization in ROBs and bone formation in growing rats, which involve osteoblast primary cilia length extension and PKA activation.


Subject(s)
Cell Differentiation/physiology , Cilia/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Electromagnetic Fields , Osteoblasts/physiology , Osteogenesis/physiology , Animals , Animals, Newborn , Capillaries/cytology , Capillaries/physiology , Cells, Cultured , Enzyme Activation/physiology , Female , Rats , Rats, Wistar , Skull/cytology , Skull/physiology
17.
Molecules ; 26(4)2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33562825

ABSTRACT

The aim is to evaluate the effects of photobiomodulation therapy (PBMT) on the guided bone regeneration process (GBR) in defects in the calvaria of rats filled with biphasic calcium phosphate associated with fibrin biopolymer. Thirty male Wistar rats were randomly separated: BMG (n = 10), defects filled with biomaterial and covered by membrane; BFMG (n = 10), biomaterial and fibrin biopolymer covered by membrane; and BFMLG (n = 10), biomaterial and fibrin biopolymer covered by membrane and biostimulated with PBMT. The animals were euthanized at 14 and 42 days postoperatively. Microtomographically, in 42 days, there was more evident bone growth in the BFMLG, limited to the margins of the defect with permanence of the particles. Histomorphologically, an inflammatory infiltrate was observed, which regressed with the formation of mineralized bone tissue. In the quantification of bone tissue, all groups had a progressive increase in new bone tissue with a significant difference in which the BFMLG showed greater bone formation in both periods (10.12 ± 0.67 and 13.85 ± 0.54), followed by BFMG (7.35 ± 0.66 and 9.41 ± 0.84) and BMG (4.51 ± 0.44 and 7.11 ± 0.44). Picrosirius-red staining showed greater birefringence of collagen fibers in yellow-green color in the BFMLG, showing more advanced bone maturation. PBMT showed positive effects capable of improving and accelerating the guided bone regeneration process when associated with biphasic calcium phosphate and fibrin biopolymer.


Subject(s)
Bone Regeneration/drug effects , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Fibrin/chemistry , Guided Tissue Regeneration/methods , Low-Level Light Therapy , Animals , Rats , Skull/cytology , Skull/drug effects , Skull/physiology
18.
J Biomed Mater Res B Appl Biomater ; 109(10): 1426-1435, 2021 10.
Article in English | MEDLINE | ID: mdl-33484103

ABSTRACT

Autologous bone grafting remains the gold standard for almost all bone void-filling orthopedic surgery. However, autologous bone grafting has several limitations, thus scientists are trying to identify an ideal synthetic material as an alternative bone graft substitute. Magnesium-doped biphasic calcium phosphate (Mg-BCP) has recently been in the spotlight and is considered to be a potential bone substitute. The Mg-BCP is a mixture of two bioceramics, that is, hydroxyapatite (HA) and ß-tricalcium phosphate (ß-TCP), doped with Mg2+ , and can be synthesized through chemical wet-precipitation, sol-gel, single diffusion gel, and solid state reactions. Regardless of the synthesis routes, it is found that the Mg2+ preferentially accommodates in ß-TCP lattice instead of the HA lattice. The addition of Mg2+ to BCP leads to desirable physicochemical properties and is found to enhance the apatite-forming ability as compared to pristine BCP. In vitro results suggest that the Mg-BCP is bioactive and not toxic to cells. Implantation of Mg-BCP in in vivo models further affirmed its biocompatibility and efficacy as a bone substitute. However, like the other bioceramics, the optimum physicochemical properties of the Mg-BCP scaffold have yet to be determined. Further investigations are required regarding Mg-BCP applications in bone tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Bone Substitutes/chemistry , Hydroxyapatites/chemistry , Magnesium/chemistry , Animals , Apatites/chemistry , Bone Regeneration , Bone and Bones/chemistry , Cations, Divalent/chemistry , Cell Adhesion , Cell Proliferation , Humans , Macrophages/cytology , Mesenchymal Stem Cells/cytology , Rats, Wistar , Skull/cytology
19.
Dev Biol ; 471: 119-137, 2021 03.
Article in English | MEDLINE | ID: mdl-33316258

ABSTRACT

Diversity of neural crest derivatives has been studied with a variety of approaches during embryonic development. In mammals Cre-LoxP lineage tracing is a robust means to fate map neural crest relying on cre driven from regulatory elements of early neural crest genes. Sox10 is an essential transcription factor for normal neural crest development. A variety of efforts have been made to label neural crest derivatives using partial Sox10 regulatory elements to drive cre expression. To date published Sox10-cre lines have focused primarily on lineage tracing in specific tissues or during early fetal development. We describe two new Sox10-cre BAC transgenes, constitutive (cre) and inducible (cre/ERT2), that contain the complete repertoire of Sox10 regulatory elements. We present a thorough expression profile of each, identifying a few novel sites of Sox10 expression not captured by other neural crest cre drivers. Comparative mapping of expression patterns between the Sox10-cre and Sox10-cre/ERT2 transgenes identified a narrow temporal window in which Sox10 expression is present in mesenchymal derivatives prior to becoming restricted to neural elements during embryogenesis. In more caudal structures, such as the intestine and lower urinary tract, our Sox10-cre BAC transgene appears to be more efficient in labeling neural crest-derived cell types than Wnt1-cre. The analysis reveals consistent expression of Sox10 in non-neural crest derived glandular epithelium, including salivary, mammary, and urethral glands of adult mice. These Sox10-cre and Sox10-cre/ERT2 transgenic lines are verified tools that will enable refined temporal and cell-type specific lineage analysis of neural crest derivatives as well as glandular tissues that rely on Sox10 for proper development and function.


Subject(s)
Gene Expression Regulation, Developmental , Mesoderm/embryology , Neural Crest/embryology , SOXE Transcription Factors/biosynthesis , Skull/embryology , Transgenes , Animals , Mesoderm/cytology , Mice , Mice, Transgenic , Neural Crest/cytology , SOXE Transcription Factors/genetics , Skull/cytology
20.
J Biomed Mater Res A ; 109(5): 788-803, 2021 05.
Article in English | MEDLINE | ID: mdl-32720351

ABSTRACT

Reconstruction of organ-specific architecture is necessary to recover the original organ function. The anisotropic structure of bone tissue is strongly related to the collagen fibril alignment and bone apatite crystal direction. Bone regeneration indicates following two main process; first, restoration of bone mineral density (BMD; bone quantity), and second, restoring bone apatite c-axis orientation (bone quality). In addition to BMD, bone quality is the most important factor among bone mechanical properties. Recovery of the original bone function requires development of novel scaffolds with simultaneous reconstruction of bone quality and quantity. Herein, novel orthophosphosilicate glass (PSG)/poly(lactic acid) composite anisotropic scaffolds were developed to control cell alignment and enhance bone formation, which are important for the simultaneous reconstruction of bone quality and quantity. The strategy to control cell alignment and bone formation involved designing anisotropic scaffolds in combination with the release of therapeutic ions by PSGs. The morphology of fibrous scaffolds containing PSGs was quantitatively designed using electrospinning. This successfully modulated cell alignment and subsequent bone apatite c-axis orientation along the fiber-oriented direction. The released silicate and Mg2+ ions from PSGs in scaffolds improved cell adhesion, proliferation, and calcification. To best of our knowledge, this is the first report demonstrating that the anisotropic scaffolds containing bioactive glasses regenerate bone tissues with simultaneous reconstruction of bone quality and quantity via stimulating osteoblasts by inorganic ions and designing morphology of scaffolds.


Subject(s)
Bone Regeneration , Glass , Polyesters , Tissue Scaffolds , Animals , Animals, Newborn , Anisotropy , Apatites/metabolism , Calcification, Physiologic , Cations , Cells, Cultured , Glass/chemistry , Materials Testing , Mice , Mice, Inbred C57BL , Nuclear Magnetic Resonance, Biomolecular , Osteoblasts/metabolism , Osteoblasts/ultrastructure , Silicates , Skull/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...