Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Bone Miner Res ; 39(2): 150-160, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38477776

ABSTRACT

Skeletal stem and progenitor cells (SSPCs) are crucial for bone development, homeostasis, and repair. SSPCs are considered to reside in a rather hypoxic niche in the bone, but distinct SSPC niches have been described in different skeletal regions, and they likely differ in oxygen and nutrient availability. Currently it remains unknown whether the different SSPC sources have a comparable metabolic profile and respond in a similar manner to hypoxia. In this study, we show that cell proliferation of all SSPCs was increased in hypoxia, suggesting that SSPCs can indeed function in a hypoxic niche in vivo. In addition, low oxygen tension increased glucose consumption and lactate production, but affected pyruvate metabolism cell-specifically. Hypoxia decreased tricarboxylic acid (TCA) cycle anaplerosis and altered glucose entry into the TCA cycle from pyruvate dehydrogenase to pyruvate carboxylase and/or malic enzyme. Finally, a switch from glutamine oxidation to reductive carboxylation was observed in hypoxia, as well as cell-specific adaptations in the metabolism of other amino acids. Collectively, our findings show that SSPCs from different skeletal locations proliferate adequately in hypoxia by rewiring glucose and amino acid metabolism in a cell-specific manner.


Skeletal stem and progenitor cells provide a lifelong cell source for bone-forming osteoblasts and these cells reside in unique microenvironments in different regions of the bone, often characterized by low oxygen levels. It was still unknown whether these regional differences resulted in diverse metabolic profiles. In this study, we show that all types of skeletal stem and progenitor cells can proliferate in low oxygen levels by adapting their metabolism of glucose and amino acids, but they differ in how they modify pyruvate metabolism.


Subject(s)
Glucose , Glutamine , Pyruvic Acid , Stem Cells , Glucose/metabolism , Glutamine/metabolism , Animals , Pyruvic Acid/metabolism , Stem Cells/metabolism , Cell Proliferation , Cell Hypoxia , Mice , Bone and Bones/metabolism , Citric Acid Cycle
2.
Nat Metab ; 6(1): 141-152, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38200114

ABSTRACT

Bone-resorbing osteoclasts are vital for postnatal bone health, as increased differentiation or activity results in skeletal pathologies such as osteoporosis. The metabolism of mature osteoclasts differs from their progenitor cells, but whether the observed metabolic changes are secondary to the altered cell state or actively drive the process of cell differentiation is unknown. Here, we show that transient activation of the serine synthesis pathway (SSP) is essential for osteoclastogenesis, as deletion of the rate-limiting enzyme phosphoglycerate dehydrogenase in osteoclast progenitors impairs their differentiation and results in increased bone mass. In addition, pharmacological phosphoglycerate dehydrogenase inhibition abrogated bone loss in a mouse model of postmenopausal osteoporosis by blocking bone resorption. Mechanistically, SSP-derived α-ketoglutarate is necessary for histone demethylases that remove repressive histone methylation marks at the nuclear factor of activated T cells, cytoplasmic 1 (Nfatc1) gene locus, thereby inducing NFATc1 expression and consequent osteoclast maturation. Taken together, this study reveals a metabolic-epigenetic coupling mechanism that directs osteoclast differentiation and suggests that the SSP can be therapeutically targeted to prevent osteoporotic bone loss.


Subject(s)
Epigenesis, Genetic , NFATC Transcription Factors , Osteoclasts , Animals , Mice , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Phosphoglycerate Dehydrogenase/genetics , Serine/genetics , Serine/metabolism
3.
Front Endocrinol (Lausanne) ; 13: 930358, 2022.
Article in English | MEDLINE | ID: mdl-35979436

ABSTRACT

Skeletal stem and progenitor cells (SSPCs) constitute a reservoir of bone-forming cells necessary for bone development, modeling and remodeling, as well as for fracture healing. Recent advances in tools to identify and isolate SSPCs have revealed that cells with multipotent properties are present not only in neonatal bone, but also in adult bone marrow and periosteum. The long bone metaphysis and endosteum have been proposed as an additional SSPC niche, although in vitro approaches to study their cellular and molecular characteristics are still limited. Here, we describe a comprehensive procedure to isolate and culture SSPCs derived from the metaphysis and endosteum of young-adult mice. Based on flow cytometry analysis of known SSPC markers, we found the presence of putative multipotent SSPCs, similar to neonatal bone tissue. In vitro, metaphyseal/endosteal SSPCs possess self-renewing capacity, and their multipotency is underscored by the ability to differentiate into the osteogenic and adipogenic lineage, while chondrogenic potential is limited. Expansion of metaphyseal/endosteal SSPCs under low oxygen conditions increases their proliferation capacity, while progenitor properties are maintained, likely reflecting their hypoxic niche in vivo. Collectively, we propose a validated isolation and culture protocol to study metaphyseal/endosteal SSPC biology in vitro.


Subject(s)
Bone and Bones , Osteogenesis , Animals , Cell Differentiation , Fracture Healing , Mice , Stem Cells
4.
Bone Res ; 10(1): 14, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35165259

ABSTRACT

The majority of the mammalian skeleton is formed through endochondral ossification starting from a cartilaginous template. Cartilage cells, or chondrocytes, survive, proliferate and synthesize extracellular matrix in an avascular environment, but the metabolic requirements for these anabolic processes are not fully understood. Here, using metabolomics analysis and genetic in vivo models, we show that maintaining intracellular serine homeostasis is essential for chondrocyte function. De novo serine synthesis through phosphoglycerate dehydrogenase (PHGDH)-mediated glucose metabolism generates nucleotides that are necessary for chondrocyte proliferation and long bone growth. On the other hand, dietary serine is less crucial during endochondral bone formation, as serine-starved chondrocytes compensate by inducing PHGDH-mediated serine synthesis. Mechanistically, this metabolic flexibility requires ATF4, a transcriptional regulator of amino acid metabolism and stress responses. We demonstrate that both serine deprivation and PHGDH inactivation enhance ATF4 signaling to stimulate de novo serine synthesis and serine uptake, respectively, and thereby prevent intracellular serine depletion and chondrocyte dysfunction. A similar metabolic adaptability between serine uptake and de novo synthesis is observed in the cartilage callus during fracture repair. Together, the results of this study reveal a critical role for PHGDH-dependent serine synthesis in maintaining intracellular serine levels under physiological and serine-limited conditions, as adequate serine levels are necessary to support chondrocyte proliferation during endochondral ossification.

5.
Dev Cell ; 53(5): 530-544.e8, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32470321

ABSTRACT

Correct functioning of chondrocytes is crucial for long bone growth and fracture repair. These cells are highly anabolic but survive and function in an avascular environment, implying specific metabolic requirements that are, however, poorly characterized. Here, we show that chondrocyte identity and function are closely linked with glutamine metabolism in a feedforward process. The master chondrogenic transcription factor SOX9 stimulates glutamine metabolism by increasing glutamine consumption and levels of glutaminase 1 (GLS1), a rate-controlling enzyme in this pathway. Consecutively, GLS1 action is critical for chondrocyte properties and function via a tripartite mechanism. First, glutamine controls chondrogenic gene expression epigenetically through glutamate dehydrogenase-dependent acetyl-CoA synthesis, necessary for histone acetylation. Second, transaminase-mediated aspartate synthesis supports chondrocyte proliferation and matrix synthesis. Third, glutamine-derived glutathione synthesis avoids harmful reactive oxygen species accumulation and allows chondrocyte survival in the avascular growth plate. Collectively, our study identifies glutamine as a metabolic regulator of cartilage fitness during bone development.


Subject(s)
Chondrocytes/metabolism , Glutamine/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/physiology , Female , Glutaminase/metabolism , Male , Mice , SOX9 Transcription Factor/metabolism
6.
Bone ; 133: 115259, 2020 04.
Article in English | MEDLINE | ID: mdl-32036051

ABSTRACT

The periosteum is critical for bone repair and contains skeletal stem cells (SSCs), but these cells are still poorly characterized. In the bone marrow, cells expressing the Nes-GFP transgene have been described to be SSCs. Here, we investigated whether Nes-GFP expression also typifies SSCs in the periosteum. We show that in adult mice, Nes-GFP cells are present in the periosteum and localize closely to blood vessels, but periosteal Nes-GFP cells express SSC and progenitor markers differently compared to Nes-GFP cells in the bone marrow. Periosteal Nes-GFP cells show in vitro clonogenicity and tri-lineage differentiation potential and they can form bone in vivo. Shortly after fracture, they start to proliferate and they contribute to the osteoblast pool during the repair process. However, periosteal Nes-GFP cells are not slow dividing nor self-renewing in vivo. These results indicate that in adult mice, periosteal Nes-GFP expressing cells are skeletal progenitors rather than true SSCs, and they participate in the fracture healing process.


Subject(s)
Osteoblasts , Periosteum , Animals , Mice , Nestin/genetics , Stem Cells , Transgenes
7.
JBMR Plus ; 2(2): 92-102, 2018 Mar.
Article in English | MEDLINE | ID: mdl-30283894

ABSTRACT

Tissue engineering is a promising branch of regenerative medicine, but its clinical application remains limited because thorough knowledge of the in vivo repair processes in these engineered implants is limited. Common techniques to study the different phases of bone repair in mice are destructive and thus not optimal to gain insight into the dynamics of this process. Instead, multiphoton-intravital microscopy (MP-IVM) allows visualization of (sub)cellular processes at high resolution and frequency over extended periods of time when combined with an imaging window that permits optical access to implants in vivo. In this study, we have developed and validated an ectopic imaging window that can be placed over a tissue-engineered construct implanted in mice. This approach did not interfere with the biological processes of bone regeneration taking place in these implants, as evidenced by histological and micro-computed tomography (µCT)-based comparison to control ectopic implants. The ectopic imaging window permitted tracking of individual cells over several days in vivo. Furthermore, the use of fluorescent reporters allowed visualization of the onset of angiogenesis and osteogenesis in these constructs. Taken together, this novel imaging window will facilitate further analysis of the spatiotemporal regulation of cellular processes in bone tissue-engineered implants and provides a powerful tool to enhance the therapeutic potential of bone tissue engineering.

8.
Nat Commun ; 9(1): 2557, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29967369

ABSTRACT

Preservation of bone mass is crucial for healthy ageing and largely depends on adequate responses of matrix-embedded osteocytes. These cells control bone formation and resorption concurrently by secreting the WNT/ß-catenin antagonist sclerostin (SOST). Osteocytes reside within a low oxygen microenvironment, but whether and how oxygen sensing regulates their function remains elusive. Here, we show that conditional deletion of the oxygen sensor prolyl hydroxylase (PHD) 2 in osteocytes results in a high bone mass phenotype, which is caused by increased bone formation and decreased resorption. Mechanistically, enhanced HIF-1α signalling increases Sirtuin 1-dependent deacetylation of the Sost promoter, resulting in decreased sclerostin expression and enhanced WNT/ß-catenin signalling. Additionally, genetic ablation of PHD2 in osteocytes blunts osteoporotic bone loss induced by oestrogen deficiency or mechanical unloading. Thus, oxygen sensing by PHD2 in osteocytes negatively regulates bone mass through epigenetic regulation of sclerostin and targeting PHD2 elicits an osteo-anabolic response in osteoporotic models.


Subject(s)
Glycoproteins/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Osteocytes/metabolism , Osteoporosis/genetics , Oxygen/metabolism , Acetylation , Adaptor Proteins, Signal Transducing , Animals , Bone Density/genetics , Carbazoles/pharmacology , Cell Line , Coculture Techniques , Disease Models, Animal , Epigenesis, Genetic/physiology , Female , Glycoproteins/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Intercellular Signaling Peptides and Proteins , Male , Mice, Inbred C57BL , Mice, Transgenic , Osteogenesis/genetics , Osteoporosis/etiology , Osteoporosis/pathology , Primary Cell Culture , Promoter Regions, Genetic , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism , Wnt Signaling Pathway/physiology
9.
Bone ; 105: 154-162, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28863946

ABSTRACT

To improve bone healing or regeneration more insight in the fate and role of the different skeletal cell types is required. Mouse models for fate mapping and lineage tracing of skeletal cells, using stage-specific promoters, have advanced our understanding of bone development, a process that is largely recapitulated during bone repair. However, validation of these models is often only performed during development, whereas proof of the activity and specificity of the used promoters during the bone regenerative process is limited. Here, we show that the regulatory elements of the 6kb collagen type II promoter are not adequate to drive gene expression during bone repair. Similarly, the 2.3kb promoter of collagen type I lacks activity in adult mice, but the 3.2kb promoter is suitable. Furthermore, Cre-mediated fate mapping allows the visualization of progeny, but this label retention may hinder to distinguish these cells from ones with active expression of the marker at later time points. Together, our results show that the lineage-specific regulatory elements driving gene expression during bone development differ from those required later in life and during bone repair, and justify validation of lineage-specific cell tracing and gene silencing strategies during fracture healing and bone regenerative applications.


Subject(s)
Aging/genetics , Bone Development/genetics , Bone and Bones/physiology , Cell Lineage/genetics , Gene Expression Regulation, Developmental , Regulatory Sequences, Nucleic Acid/genetics , Animals , Bony Callus/pathology , Cartilage/growth & development , Chondrogenesis/genetics , Collagen Type II/genetics , Green Fluorescent Proteins/metabolism , Integrases/metabolism , Mice , Osteogenesis/genetics , Promoter Regions, Genetic , Reproducibility of Results , Transcription, Genetic , Transgenes , Wound Healing/genetics
10.
Stem Cells ; 32(9): 2407-18, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24989687

ABSTRACT

The preservation of the bone-forming potential of skeletal progenitor cells during their ex vivo expansion remains one of the major challenges for cell-based bone regeneration strategies. We report that expansion of murine periosteal cells in the presence of FGF2, a signal present during the early stages of fracture healing, is necessary and sufficient to maintain their ability to organize in vivo into a cartilage template which gives rise to mature bone. Implantation of FGF2-primed cells in a large bone defect in mice resulted in complete healing, demonstrating the feasibility of using this approach for bone tissue engineering purposes. Mechanistically, the enhanced endochondral ossification potential of FGF2-expanded periosteal cells is predominantly driven by an increased production of BMP2 and is additionally linked to an improved preservation of skeletal progenitor cells in the cultures. This characteristic is unique for periosteal cells, as FGF2-primed bone marrow stromal cells formed significantly less bone and progressed exclusively through the intramembranous pathway, revealing essential differences between both cell pools. Taken together, our findings provide insight in the molecular regulation of fracture repair by identifying a unique interaction between periosteal cells and FGF2. These insights may promote the development of cell-based therapeutic strategies for bone regeneration which are independent of the in vivo use of growth factors, thus limiting undesired side effects.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Periosteum/cytology , Stem Cells/cytology , Tissue Engineering/methods , Animals , Bone Morphogenetic Protein 2/genetics , Cell Culture Techniques , Gene Expression , Mice , Mice, Inbred C57BL , Periosteum/drug effects , Periosteum/metabolism , Stem Cells/drug effects
11.
EMBO J ; 29(2): 424-41, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20010698

ABSTRACT

Vascular endothelial growth factor (VEGF) and beta-catenin both act broadly in embryogenesis and adulthood, including in the skeletal and vascular systems. Increased or deregulated activity of these molecules has been linked to cancer and bone-related pathologies. By using novel mouse models to locally increase VEGF levels in the skeleton, we found that embryonic VEGF over-expression in osteo-chondroprogenitors and their progeny largely pheno-copied constitutive beta-catenin activation. Adult induction of VEGF in these cell populations dramatically increased bone mass, associated with aberrant vascularization, bone marrow fibrosis and haematological anomalies. Genetic and pharmacological interventions showed that VEGF increased bone mass through a VEGF receptor 2- and phosphatidyl inositol 3-kinase-mediated pathway inducing beta-catenin transcriptional activity in endothelial and osteoblastic cells, likely through modulation of glycogen synthase kinase 3-beta phosphorylation. These insights into the actions of VEGF in the bone and marrow environment underscore its power as pleiotropic bone anabolic agent but also warn for caution in its therapeutic use. Moreover, the finding that VEGF can modulate beta-catenin activity may have widespread physiological and clinical ramifications.


Subject(s)
Bone and Bones/metabolism , Bone and Bones/pathology , Gene Expression Regulation, Developmental , Vascular Endothelial Growth Factor A/metabolism , beta Catenin/metabolism , Animals , Bone and Bones/embryology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Endothelial Cells/cytology , Humans , Mesoderm/cytology , Mice , Mice, Transgenic , Morphogenesis , Osteoblasts/cytology , Phosphatidylinositol 3-Kinases/metabolism , Stem Cells/cytology , Stromal Cells/cytology , Vascular Endothelial Growth Factor A/genetics , beta Catenin/genetics
12.
Arch Biochem Biophys ; 460(2): 300-5, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17224125

ABSTRACT

Reduced intestinal calcium absorption may be part of the pathogenesis of glucocorticoid-induced osteoporosis. 1,25(OH)2D3 is the major regulator of the expression of the active duodenal calcium absorption genes: TRPV6 (influx), calbindin-D9K (intracellular transfer) and PMCA1b (extrusion). We investigated the influence of dexamethasone (5 days: 2 mg/kg bw) on calcium absorption in vivo and on the expression of intestinal and renal calcium transporters in calcium-deprived mice. Total and free 1,25(OH)2D3-concentrations were halved, in line with decreased 25(OH)D3-1-alpha-hydroxylase and increased 24-hydroxylase expression. Nevertheless, no difference in duodenal or renal calcium transporter expression pattern could be detected between vehicle and dexamethasone-treated mice. Accordingly, dexamethasone did not affect in vivo calcium absorption. By contrast, increased calcemia and collagen C-terminal telopeptide levels reflected increased bone resorption. Decreased osteocalcin levels suggested impaired bone formation. Hence, short-term glucocorticoid excess in young animals affected bone metabolism without detectable changes in intestinal or renal calcium handling.


Subject(s)
Calcitriol/metabolism , Calcium/metabolism , Dexamethasone/adverse effects , Duodenum/metabolism , Glucocorticoids/adverse effects , Osteoporosis/metabolism , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/biosynthesis , Absorption/drug effects , Animals , Bone Resorption/chemically induced , Bone Resorption/metabolism , Calcium/deficiency , Calcium-Transporting ATPases/metabolism , Dexamethasone/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glucocorticoids/pharmacology , Male , Mice , Osteocalcin/metabolism , Osteoporosis/chemically induced
13.
J Clin Invest ; 116(12): 3150-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17099775

ABSTRACT

Genomic actions induced by 1alpha25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] are crucial for normal bone metabolism, mainly because they regulate active intestinal calcium transport. To evaluate whether the vitamin D receptor (VDR) has a specific role in growth-plate development and endochondral bone formation, we investigated mice with conditional inactivation of VDR in chondrocytes. Growth-plate chondrocyte development was not affected by the lack of VDR. Yet vascular invasion was impaired, and osteoclast number was reduced in juvenile mice, resulting in increased trabecular bone mass. In vitro experiments confirmed that VDR signaling in chondrocytes directly regulated osteoclastogenesis by inducing receptor activator of NF-kappaB ligand (RANKL) expression. Remarkably, mineral homeostasis was also affected in chondrocyte-specific VDR-null mice, as serum phosphate and 1,25(OH)(2)D levels were increased in young mice, in whom growth-plate activity is important. Both in vivo and in vitro analysis indicated that VDR inactivation in chondrocytes reduced the expression of FGF23 by osteoblasts and consequently led to increased renal expression of 1alpha-hydroxylase and of sodium phosphate cotransporter type IIa. Taken together, our findings provide evidence that VDR signaling in chondrocytes is required for timely osteoclast formation during bone development and for the endocrine action of bone in phosphate homeostasis.


Subject(s)
Chondrocytes/metabolism , Fibroblast Growth Factors/genetics , Osteoblasts/metabolism , Receptors, Calcitriol/physiology , Animals , Animals, Newborn , Bone Development/genetics , Bone Development/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Chondrocytes/cytology , Fibroblast Growth Factor-23 , Gene Expression/genetics , Growth Plate/cytology , Growth Plate/metabolism , Homeostasis/physiology , Immunohistochemistry , Mice , Mutation/genetics , Osteoblasts/cytology , Osteoclasts/cytology , Osteoclasts/metabolism , Osteogenesis/genetics , Osteogenesis/physiology , Phosphates/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/analysis , RANK Ligand/genetics , RANK Ligand/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Vitamin D/analogs & derivatives , Vitamin D/genetics , Vitamin D/physiology
14.
J Clin Invest ; 116(5): 1230-42, 2006 May.
Article in English | MEDLINE | ID: mdl-16614757

ABSTRACT

Current therapies for delayed- or nonunion bone fractures are still largely ineffective. Previous studies indicated that the VEGF homolog placental growth factor (PlGF) has a more significant role in disease than in health. Therefore we investigated the role of PlGF in a model of semi-stabilized bone fracture healing. Fracture repair in mice lacking PlGF was impaired and characterized by a massive accumulation of cartilage in the callus, reminiscent of delayed- or nonunion fractures. PlGF was required for the early recruitment of inflammatory cells and the vascularization of the fracture wound. Interestingly, however, PlGF also played a role in the subsequent stages of the repair process. Indeed in vivo and in vitro findings indicated that PlGF induced the proliferation and osteogenic differentiation of mesenchymal progenitors and stimulated cartilage turnover by particular MMPs. Later in the process, PlGF was required for the remodeling of the newly formed bone by stimulating osteoclast differentiation. As PlGF expression was increased throughout the process of bone repair and all the important cell types involved expressed its receptor VEGFR-1, the present data suggest that PlGF is required for mediating and coordinating the key aspects of fracture repair. Therefore PlGF may potentially offer therapeutic advantages for fracture repair.


Subject(s)
Bone Remodeling , Cartilage/cytology , Fracture Healing , Mesoderm/cytology , Pregnancy Proteins/physiology , Animals , Cartilage/metabolism , Cell Differentiation , Cell Proliferation , Inflammation , Mice , Mice, Transgenic , Models, Biological , Osteoclasts/cytology , Placenta Growth Factor , Vascular Endothelial Growth Factor Receptor-1/metabolism
15.
J Clin Invest ; 113(2): 188-99, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14722611

ABSTRACT

VEGF is crucial for metaphyseal bone vascularization. In contrast, the angiogenic factors required for vascularization of epiphyseal cartilage are unknown, although this represents a developmentally and clinically important aspect of bone growth. The VEGF gene is alternatively transcribed into VEGF(120), VEGF(164), and VEGF(188) isoforms that differ in matrix association and receptor binding. Their role in bone development was studied in mice expressing single isoforms. Here we report that expression of only VEGF(164) or only VEGF(188) (in VEGF(188/188) mice) was sufficient for metaphyseal development. VEGF(188/188) mice, however, showed dwarfism, disrupted development of growth plates and secondary ossification centers, and knee joint dysplasia. This phenotype was at least partly due to impaired vascularization surrounding the epiphysis, resulting in ectopically increased hypoxia and massive chondrocyte apoptosis in the interior of the epiphyseal cartilage. In addition to the vascular defect, we provide in vitro evidence that the VEGF(188) isoform alone is also insufficient to regulate chondrocyte proliferation and survival responses to hypoxia. Consistent herewith, chondrocytes in or close to the hypoxic zone in VEGF(188/188) mice showed increased proliferation and decreased differentiation. These findings indicate that the insoluble VEGF(188) isoform is insufficient for establishing epiphyseal vascularization and regulating cartilage development during endochondral bone formation.


Subject(s)
Chondrocytes/cytology , Epiphyses/blood supply , Epiphyses/metabolism , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/genetics , Xenopus Proteins , Angiography , Animals , Bone Development , Bromodeoxyuridine/pharmacology , Cartilage/cytology , Cartilage/pathology , Cell Differentiation , Cell Division , Cell Survival , Chondrocytes/metabolism , DNA, Complementary/metabolism , Hypoxia , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Models, Biological , Mutagenesis , Neovascularization, Physiologic , Nerve Tissue Proteins/metabolism , Phenotype , Protein Binding , Protein Isoforms , RNA, Messenger/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Recombination, Genetic , Ribonucleoproteins/metabolism
16.
J Cell Biol ; 162(6): 1017-29, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12963707

ABSTRACT

Here, we report on the identification of nucleolar spindle-associated protein (NuSAP), a novel 55-kD vertebrate protein with selective expression in proliferating cells. Its mRNA and protein levels peak at the transition of G2 to mitosis and abruptly decline after cell division. Microscopic analysis of both fixed and live mammalian cells showed that NuSAP is primarily nucleolar in interphase, and localizes prominently to central spindle microtubules during mitosis. Direct interaction of NuSAP with microtubules was demonstrated in vitro. Overexpression of NuSAP caused profound bundling of cytoplasmic microtubules in interphase cells, and this relied on a COOH-terminal microtubule-binding domain. In contrast, depletion of NuSAP by RNA interference resulted in aberrant mitotic spindles, defective chromosome segregation, and cytokinesis. In addition, many NuSAP-depleted interphase cells had deformed nuclei. Both overexpression and knockdown of NuSAP impaired cell proliferation. These results suggest a crucial role for NuSAP in spindle microtubule organization.


Subject(s)
Cell Nucleolus/metabolism , Microtubule-Associated Proteins/metabolism , Mitosis/genetics , Nuclear Proteins/metabolism , Spindle Apparatus/metabolism , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , Cell Line , Cell Nucleolus/genetics , Cell Nucleus/genetics , Cell Nucleus/pathology , Chromosome Segregation/genetics , DNA, Complementary/analysis , Eukaryotic Cells/metabolism , G2 Phase/genetics , Mice , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Molecular Sequence Data , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Protein Binding/genetics , Protein Structure, Tertiary/genetics , RNA Interference , RNA, Messenger/metabolism , Spindle Apparatus/genetics , Up-Regulation/drug effects , Up-Regulation/physiology
17.
Pediatr Res ; 54(4): 466-73, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12815117

ABSTRACT

Fetal mineralization appears to be driven by the pregnancy-induced stimulation of intestinal Ca absorption. We thus hypothesized that mineralization would be impaired in fetuses of mice that lack the vitamin D receptor (VDR). Here we report on the maternal response to pregnancy, and the fetal mineralization, in mice with a homozygous disruption of the VDR gene (VDR-/-) mated with wild-type (wt) males. We found that VDR-/- mice show mild hypocalcemia, clear rickets and osteomalacia on bone histomorphometry, lower cortical bone density on quantitative tomography, and reduced concentrations of calbindin-D9k (CaBP-D9k) in duodenal mucosa and kidney. The skeletal response to pregnancy was comparable in wt and VDR-/- mice; duodenal CaBP-D9k concentrations increased during pregnancy in VDR-/- as in wt mice, but remained 40% lower than in wt mice. We confirmed our hypothesis that mineralization is defective in d18.5 VDR+/- fetuses of VDR-/- mice, both by whole-body Ca determination and histomorphometric evaluation; the number of osteoclastic cells in bone was increased. The fetuses were hypercalcemic and had a 5-fold increase in circulating 1,25(OH)2D3. We then studied pregnancies in VDR-/- females, mated with wt males, fed a high Ca/P/lactose rescue diet during pregnancy. The rescue diet normalized the mineralization, the number of osteoclastic cells, and plasma Ca and 1,25(OH)2D3 concentrations in the fetuses. We interpret the data as evidence that, to ensure normal fetal mineralization, the maternal VDR-dependent intestinal Ca absorption can be substituted by passive Ca absorption entrained by a higher Ca intake. Alternatively or additionally, elevated 1,25(OH)2D3 in utero may disturb bone development.


Subject(s)
Bone and Bones/metabolism , Bone and Bones/physiopathology , Calcification, Physiologic/physiology , Calcium/administration & dosage , Fetus/physiology , Pregnancy, Animal , Receptors, Calcitriol/metabolism , Animals , Body Weight , Bone and Bones/pathology , Calbindins , Calcium/metabolism , Cholecalciferol/blood , Dietary Supplements , Duodenum/metabolism , Female , Kidney/metabolism , Male , Mice , Mice, Knockout , Phenotype , Placenta/anatomy & histology , Placenta/metabolism , Pregnancy , Receptors, Calcitriol/genetics , S100 Calcium Binding Protein G/metabolism
18.
Mech Dev ; 111(1-2): 61-73, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11804779

ABSTRACT

Vascular endothelial growth factor (VEGF)-mediated angiogenesis is an important part of bone formation. To clarify the role of VEGF isoforms in endochondral bone formation, we examined long bone development in mice expressing exclusively the VEGF120 isoform (VEGF120/120 mice). Neonatal VEGF120/120 long bones showed a completely disturbed vascular pattern, concomitant with a 35% decrease in trabecular bone volume, reduced bone growth and a 34% enlargement of the hypertrophic chondrocyte zone of the growth plate. Surprisingly, embryonic hindlimbs at a stage preceding capillary invasion exhibited a delay in bone collar formation and hypertrophic cartilage calcification. Expression levels of marker genes of osteoblast and hypertrophic chondrocyte differentiation were significantly decreased in VEGF120/120 bones. Furthermore, inhibition of all VEGF isoforms in cultures of embryonic cartilaginous metatarsals, through the administration of a soluble receptor chimeric protein (mFlt-1/Fc), retarded the onset and progression of ossification, suggesting that osteoblast and/or hypertrophic chondrocyte development were impaired. The initial invasion by osteoclasts and endothelial cells into VEGF120/120 bones was retarded, associated with decreased expression of matrix metalloproteinase-9. Our findings indicate that expression of VEGF164 and/or VEGF188 is important for normal endochondral bone development, not only to mediate bone vascularization but also to allow normal differentiation of hypertrophic chondrocytes, osteoblasts, endothelial cells and osteoclasts.


Subject(s)
Bone Development , Bone and Bones/blood supply , Endothelial Growth Factors/physiology , Lymphokines/physiology , Neovascularization, Physiologic/genetics , Animals , Cartilage/abnormalities , Cartilage/embryology , Cell Differentiation/genetics , Chondrocytes/pathology , Chondrocytes/physiology , Endothelium, Vascular/pathology , Gene Deletion , Gene Expression Regulation, Developmental , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Metatarsal Bones/embryology , Metatarsal Bones/pathology , Mice , Mice, Mutant Strains , Ossification, Heterotopic , Osteoblasts/pathology , Osteoblasts/physiology , Osteoclasts/pathology , Osteoclasts/physiology , Protein Isoforms/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...