Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters










Publication year range
2.
Eur J Cell Biol ; 97(8): 568-579, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30424898

ABSTRACT

Osteoclasts are the main cells responsible for the resorption of mineralized extracellular matrices. They are the major targets for anti-resorptive therapies to manage osteoporosis, a major public health problem. Osteoclasts are giant multinucleated cells that can organize their a unique adhesion structure based on a belt of podosomes, which is the keystone of the bone resorption apparatus. We combined differential transcriptomics and siRNA screening approaches to get a broader view of cytoskeletal regulators that participate in the control of osteoclast cytoskeleton and identify novel regulators of bone resorption by osteoclasts. We identified 20 new candidate regulators of osteoclasts cytoskeleton including Fkbp15, Spire1, Tacc2 and RalA, for which we confirmed they are necessary for proper organization of the podosome belt. We also showed that Anillin, well known for its role during cytokinesis, is essential in osteoclasts for correct podosome patterning and efficient bone resorption. In particular, Anillin controls the levels of the GTPase RhoA, a known regulator of osteoclast cytoskeleton and resorption activity. Finally, we set up and validated an automated imaging strategy based on open-source software for automatic and objective measurement of actin cytoskeleton organization in osteoclasts. We provide these pipelines that are useful to automatically assess the effect of collections of siRNAs or chemical compounds on osteoclast cytoskeleton or differentiation.


Subject(s)
Actin Cytoskeleton/metabolism , Bone Resorption/metabolism , Bone Resorption/pathology , Contractile Proteins/metabolism , Imaging, Three-Dimensional , Mitosis , Osteoclasts/metabolism , RNA, Small Interfering/metabolism , Animals , Automation , Cytoskeletal Proteins/metabolism , Gene Silencing , Mice, Inbred C57BL , Podosomes/metabolism
3.
Eur J Cell Biol ; 97(8): 533-545, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30287085

ABSTRACT

Actin subunits assemble into actin filaments whose dynamics and three-dimensional architectures are further regulated by a variety of cellular factors to establish the functional actin cytoskeleton. The C-glucosidic ellagitannin vescalagin and its simpler analogue vescalin, affect both the dynamics and the ultrastructure of the actin cytoskeleton by directly binding to F-actin. Herein, we show that in vitro, the two compounds induce the formation of distinct F-actin networks characterized by different superstructures and dynamics. In living mature osteoclasts, highly specialized bone-degrading cells that constantly remodel their cytoskeleton, vescalagin and vescalin alter actin dynamics at podosomes and compromise the integrity of the podosome belt that forms the bone-degrading apparatus. Both compounds target the bone-resorbing activity at concentrations that preserve osteoclastic maturation and survival and with no detectable impact on the behaviour of bone-forming osteoblastic cells. This anti-osteoclastic activity of vescalagin and vescalin reveals the potential of targeting actin dynamics as a new therapeutic opportunity and, in this case, as a plausible approach for the local treatment of osteoporosis.


Subject(s)
Actins/metabolism , Glucosides/pharmacology , Hydrolyzable Tannins/pharmacology , Osteoclasts/cytology , Osteoclasts/metabolism , Actin Cytoskeleton/metabolism , Animals , Bone Resorption/pathology , Cell Adhesion/drug effects , Cell Differentiation , Cell Survival/drug effects , Cytoskeleton/metabolism , Extracellular Matrix/metabolism , Glucosides/chemistry , Hydrolyzable Tannins/chemistry , Mice, Inbred C57BL , Osteoclasts/drug effects , Podosomes/metabolism , Polymerization
4.
Proc Natl Acad Sci U S A ; 115(11): E2556-E2565, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29463701

ABSTRACT

Bone deficits are frequent in HIV-1-infected patients. We report here that osteoclasts, the cells specialized in bone resorption, are infected by HIV-1 in vivo in humanized mice and ex vivo in human joint biopsies. In vitro, infection of human osteoclasts occurs at different stages of osteoclastogenesis via cell-free viruses and, more efficiently, by transfer from infected T cells. HIV-1 infection markedly enhances adhesion and osteolytic activity of human osteoclasts by modifying the structure and function of the sealing zone, the osteoclast-specific bone degradation machinery. Indeed, the sealing zone is broader due to F-actin enrichment of its basal units (i.e., the podosomes). The viral protein Nef is involved in all HIV-1-induced effects partly through the activation of Src, a regulator of podosomes and of their assembly as a sealing zone. Supporting these results, Nef-transgenic mice exhibit an increased osteoclast density and bone defects, and osteoclasts derived from these animals display high osteolytic activity. Altogether, our study evidences osteoclasts as host cells for HIV-1 and their pathological contribution to bone disorders induced by this virus, in part via Nef.


Subject(s)
Bone Resorption/etiology , HIV Infections/complications , HIV-1/physiology , Osteoclasts/virology , Actins/metabolism , Animals , Bone Resorption/metabolism , Bone Resorption/pathology , Bone Resorption/physiopathology , Bone and Bones/metabolism , Cell Adhesion , Female , HIV Infections/metabolism , HIV Infections/pathology , HIV Infections/virology , HIV-1/genetics , Humans , Mice , Osteoclasts/cytology , Osteoclasts/metabolism , nef Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/metabolism
5.
PLoS One ; 10(7): e0132513, 2015.
Article in English | MEDLINE | ID: mdl-26168340

ABSTRACT

AIM: The cysteine protease cathepsin K (CatK), abundantly expressed in osteoclasts, is responsible for the degradation of bone matrix proteins, including collagen type 1. Thus, CatK is an attractive target for new anti-resorptive osteoporosis therapies, but the wider effects of CatK inhibitors on bone cells also need to be evaluated to assess their effects on bone. Therefore, we selected, among a series of synthetized isothiosemicarbazides, two molecules which are highly selective CatK inhibitors (CKIs) to test their effects on osteoblasts and osteoclasts. RESEARCH DESIGN AND METHODS: Cell viability upon treatment of CKIs were was assayed on human osteoblast-like Saos-2, mouse monocyte cell line RAW 264.7 and mature mouse osteoclasts differentiated from bone marrow. Osteoblast-induced mineralization in Saos-2 cells and in mouse primary osteoblasts from calvaria, with or without CKIs,; were was monitored by Alizarin Red staining and alkaline phosphatase activity, while osteoclast-induced bone resorption was performed on bovine slices. RESULTS: Treatments with two CKIs, CKI-8 and CKI-13 in human osteoblast-like Saos-2, murine RAW 264.7 macrophages stimulated with RANKL and mouse osteoclasts differentiated from bone marrow stimulated with RANKL and MCSF were found not to be toxic at doses of up to 100 nM. As probed by Alizarin Red staining, CKI-8 did not inhibit osteoblast-induced mineralization in mouse primary osteoblasts as well as in osteoblast-like Saos-2 cells. However, CKI-13 led to a reduction in mineralization of around 40% at 10-100 nM concentrations in osteoblast-like Saos-2 cells while it did not in primary cells. After a 48-hour incubation, both CKI-8 and CKI-13 decreased bone resorption on bovine bone slices. CKI-13 was more efficient than the commercial inhibitor E-64 in inhibiting bone resorption induced by osteoclasts on bovine bone slices. Both CKI-8 and CKI-13 created smaller bone resorption pits on bovine bone slices, suggesting that the mobility of osteoclasts was slowed down by the addition of CKI-8 and CKI-13. CONCLUSION: CKI-8 and CKI-13 screened here show promise as antiresorptive osteoporosis therapeutics but some off target effects on osteoblasts were found with CKI-13.


Subject(s)
Bone Resorption/drug therapy , Calcification, Physiologic/drug effects , Cathepsin K/antagonists & inhibitors , Osteoclasts/drug effects , Animals , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Humans , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/physiology , Osteoclasts/physiology
6.
Calcif Tissue Int ; 97(1): 69-79, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25987164

ABSTRACT

Vitamin D and FGF23 play a major role in calcium/phosphate balance. Vitamin D may control bone resorption but the potential role of FGF23 has never been evaluated. The objective of this study was therefore to compare the effects of vitamin D and FGF23 on osteoclast differentiation and activity in human monocyte-derived osteoclasts. Human monocytes, purified from blood of healthy donors, were incubated with M-CSF and RANKL to obtain mature multinucleated osteoclasts (MNC). Experiments were carried out to assess the effects of FGF23 as compared to native vitamin D (25-D) and active vitamin D (1,25-D) on osteoclast differentiation and on bone-resorbing osteoclast activity. Additional experiments with the pan fibroblast growth factor receptor inhibitor (FGFR-i) were performed. Phosphorylation Akt and Erk pathways were analyzed by Western blot analyses. Both 1,25-D and FGF23, to a lesser extent, significantly inhibited osteoclastogenesis at early stages; when adding FGFR-i, osteoclast formation was restored. Biochemical experiments showed an activation of the Akt and Erk pathways under FGF23 treatment. In contrast, in terms of activity, 1,25-D had no effect on resorption, whereas FGF23 slightly but significantly increased bone resorption; 25-D had no effects on either differentiation or on activity. These data show that 1,25-D inhibits osteoclastogenesis without regulating osteoclast-mediated bone resorption activity; FGF23 has biphasic effects on osteoclast physiology, inhibiting osteoclast formation while stimulating slightly osteoclast activity. These results may be of importance and taken into account in chronic kidney disease when therapies modulating FGF23 are available.


Subject(s)
Cell Differentiation/drug effects , Fibroblast Growth Factors/pharmacology , Osteoclasts/drug effects , Vitamin D/pharmacology , Bone Resorption/drug therapy , Cells, Cultured , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Humans , Macrophage Colony-Stimulating Factor/metabolism , Monocytes/drug effects , Monocytes/metabolism , Osteoclasts/metabolism , Vitamin D/metabolism
7.
J Infect Dis ; 211(4): 571-81, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25006047

ABSTRACT

BACKGROUND: Bone and joint infection, mainly caused by Staphylococcus aureus, is associated with significant morbidity and mortality, characterized by severe inflammation and progressive bone destruction. Studies mostly focused on the interaction between S. aureus and osteoblasts, the bone matrix-forming cells, while interactions between S. aureus and osteoclasts, the only cells known to be able to degrade bone, have been poorly explored. METHODS: We developed an in vitro infection model of primary murine osteoclasts to study the direct impact of live S. aureus on osteoclastogenesis and osteoclast resorption activity. RESULTS: Staphylococcal infection of bone marrow-derived osteoclast precursors induced their differentiation into activated macrophages that actively secreted proinflammatory cytokines. These cytokines enhanced the bone resorption capacity of uninfected mature osteoclasts and promoted osteoclastogenesis of the uninfected precursors at the site of infection. Moreover, infection of mature osteoclasts by live S. aureus directly enhanced their ability to resorb bone by promoting cellular fusion. CONCLUSIONS: Our results highlighted two complementary mechanisms involved in bone loss during bone and joint infection, suggesting that osteoclasts could be a pivotal target for limiting bone destruction.


Subject(s)
Bone Resorption/microbiology , Host-Pathogen Interactions/physiology , Osteoclasts/microbiology , Osteoclasts/physiology , Staphylococcus aureus/pathogenicity , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Cytokines/metabolism , Durapatite , Mice , Models, Biological , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics
8.
Autophagy ; 10(11): 1965-77, 2014.
Article in English | MEDLINE | ID: mdl-25484092

ABSTRACT

Bone remodeling is a tightly controlled mechanism in which osteoblasts (OB), the cells responsible for bone formation, osteoclasts (OC), the cells specialized for bone resorption, and osteocytes, the multifunctional mechanosensing cells embedded in the bone matrix, are the main actors. Increased oxidative stress in OB, the cells producing and mineralizing bone matrix, has been associated with osteoporosis development but the role of autophagy in OB has not yet been addressed. This is the goal of the present study. We first show that the autophagic process is induced in OB during mineralization. Then, using knockdown of autophagy-essential genes and OB-specific autophagy-deficient mice, we demonstrate that autophagy deficiency reduces mineralization capacity. Moreover, our data suggest that autophagic vacuoles could be used as vehicles in OB to secrete apatite crystals. In addition, autophagy-deficient OB exhibit increased oxidative stress and secretion of the receptor activator of NFKB1 (TNFSF11/RANKL), favoring generation of OC, the cells specialized in bone resorption. In vivo, we observed a 50% reduction in trabecular bone mass in OB-specific autophagy-deficient mice. Taken together, our results show for the first time that autophagy in OB is involved both in the mineralization process and in bone homeostasis. These findings are of importance for mineralized tissues which extend from corals to vertebrates and uncover new therapeutic targets for calcified tissue-related metabolic pathologies.


Subject(s)
Autophagy , Bone and Bones/metabolism , Osteoblasts/cytology , Animals , Bone Remodeling , Bone Resorption , Cell Line, Tumor , Female , Green Fluorescent Proteins/metabolism , Homeostasis , Mice , Mice, Transgenic , Microscopy, Confocal , NF-kappa B p50 Subunit/metabolism , Osteoclasts/metabolism , Oxidative Stress , RANK Ligand/metabolism , Rats , X-Ray Microtomography
9.
Cell Adh Migr ; 8(3): 191-204, 2014.
Article in English | MEDLINE | ID: mdl-24714644

ABSTRACT

Osteoclasts are the cells responsible for physiological bone resorption. A specific organization of their most prominent cytoskeletal structures, podosomes, is crucial for the degradation of mineralized bone matrix. Each podosome is constituted of an F-actin-enriched central core surrounded by a loose F-actin network, called the podosome cloud. In addition to intrinsic actin dynamics, podosomes are defined by their adhesion to the extracellular matrix, mainly via core-linking CD44 and cloud-linking integrins. These properties allow podosomes to collectively evolve into different patterns implicated in migration and bone resorption. Indeed, to resorb bone, osteoclasts polarize, actively secrete protons, and proteases into the resorption pit where these molecules are confined by a podosome-containing sealing zone. Here, we review recent advancements on podosome structure and regulatory pathways in osteoclasts. We also discuss the distinct functions of different podosome patterns during the lifespan of a single osteoclast.


Subject(s)
Bone Resorption/metabolism , Osteoclasts/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Adhesion/physiology , Cell Differentiation/physiology , Humans , Osteoclasts/cytology
10.
J Biol Chem ; 289(10): 6551-6564, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24429286

ABSTRACT

Lysophosphatidic acid (LPA) is a natural bioactive lipid that acts through six different G protein-coupled receptors (LPA1-6) with pleiotropic activities on multiple cell types. We have previously demonstrated that LPA is necessary for successful in vitro osteoclastogenesis of bone marrow cells. Bone cells controlling bone remodeling (i.e. osteoblasts, osteoclasts, and osteocytes) express LPA1, but delineating the role of this receptor in bone remodeling is still pending. Despite Lpar1(-/-) mice displaying a low bone mass phenotype, we demonstrated that bone marrow cell-induced osteoclastogenesis was reduced in Lpar1(-/-) mice but not in Lpar2(-/-) and Lpar3(-/-) animals. Expression of LPA1 was up-regulated during osteoclastogenesis, and LPA1 antagonists (Ki16425, Debio0719, and VPC12249) inhibited osteoclast differentiation. Blocking LPA1 activity with Ki16425 inhibited expression of nuclear factor of activated T-cell cytoplasmic 1 (NFATc1) and dendritic cell-specific transmembrane protein and interfered with the fusion but not the proliferation of osteoclast precursors. Similar to wild type osteoclasts treated with Ki16425, mature Lpar1(-/-) osteoclasts had reduced podosome belt and sealing zone resulting in reduced mineralized matrix resorption. Additionally, LPA1 expression markedly increased in the bone of ovariectomized mice, which was blocked by bisphosphonate treatment. Conversely, systemic treatment with Debio0719 prevented ovariectomy-induced cancellous bone loss. Moreover, intravital multiphoton microscopy revealed that Debio0719 reduced the retention of CX3CR1-EGFP(+) osteoclast precursors in bone by increasing their mobility in the bone marrow cavity. Overall, our results demonstrate that LPA1 is essential for in vitro and in vivo osteoclast activities. Therefore, LPA1 emerges as a new target for the treatment of diseases associated with excess bone loss.


Subject(s)
Bone Resorption/pathology , Membrane Proteins/metabolism , NFATC Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Osteoclasts/pathology , Receptors, Lysophosphatidic Acid/physiology , Animals , Bone Marrow Cells/pathology , Bone Resorption/drug therapy , Bone Resorption/genetics , Cell Differentiation/drug effects , Cell Movement , Female , Isoxazoles/pharmacology , Mice , Mice, Inbred BALB C , Mice, Knockout , Oleic Acids/pharmacology , Organophosphates/pharmacology , Osteoclasts/drug effects , Osteoclasts/metabolism , Propionates/pharmacology , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics
11.
J Bone Miner Res ; 29(5): 1158-69, 2014.
Article in English | MEDLINE | ID: mdl-24127173

ABSTRACT

Receptor activator of NF-κB ligand (RANKL) plays a key role in osteoclast-induced bone resorption across a range of degenerative bone diseases, and its specific inhibition has been recently approved as a treatment for women with postmenopausal osteoporosis at high or increased risk of fracture in the United States and globally. In the present study, we generated transgenic mice (TghuRANKL) carrying the human RANKL (huRANKL) genomic region and achieved a physiologically relevant pattern of RANKL overexpression in order to establish novel genetic models for assessing skeletal and extraskeletal pathologies associated with excessive RANKL and for testing clinical therapeutic candidates that inhibit human RANKL. TghuRANKL mice of both sexes developed early-onset bone loss, and the levels of huRANKL expression were correlated with bone resorption and disease severity. Low copy Tg5516 mice expressing huRANKL at low levels displayed a mild osteoporotic phenotype as shown by trabecular bone loss and reduced biomechanical properties. Notably, overexpression of huRANKL, in the medium copy Tg5519 line, resulted in severe early-onset osteoporosis characterized by lack of trabecular bone, destruction of the growth plate, increased osteoclastogenesis, bone marrow adiposity, increased bone remodeling, and severe cortical bone porosity accompanied by decreased bone strength. An even more severe skeletal phenotype developed in the high copy Tg5520 founder with extensive soft tissue calcification. Model validation was further established by evidence that denosumab, an antibody that inhibits human but not murine RANKL, fully corrected the hyper-resorptive and osteoporotic phenotypes of Tg5519 mice. Furthermore, overexpression of huRANKL rescued osteopetrotic phenotypes of RANKL-defective mice. These novel huRANKL transgenic models of osteoporosis represent an important advance for understanding the pathogenesis and treatment of high-turnover bone diseases and other disease states caused by excessive RANKL.


Subject(s)
Gene Expression Regulation , Osteoporosis/genetics , Osteoporosis/metabolism , RANK Ligand/biosynthesis , Animals , Calcinosis/genetics , Calcinosis/metabolism , Calcinosis/pathology , Disease Models, Animal , Female , Growth Plate/metabolism , Growth Plate/pathology , Humans , Mice , Mice, Transgenic , Models, Genetic , Osteoclasts/pathology , Osteopetrosis/genetics , Osteopetrosis/metabolism , Osteopetrosis/pathology , Osteoporosis/pathology , RANK Ligand/genetics
12.
Mol Biol Cell ; 25(3): 380-96, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24284899

ABSTRACT

The function of osteoclasts (OCs), multinucleated giant cells (MGCs) of the monocytic lineage, is bone resorption. To resorb bone, OCs form podosomes. These are actin-rich adhesive structures that pattern into rings that drive OC migration and into "sealing-zones" (SZs) that confine the resorption lacuna. Although changes in actin dynamics during podosome patterning have been documented, the mechanisms that regulate these changes are largely unknown. From human monocytic precursors, we differentiated MGCs that express OC degradation enzymes but are unable to resorb the mineral matrix. We demonstrated that, despite exhibiting bona fide podosomes, these cells presented dysfunctional SZs. We then performed two-step differential transcriptomic profiling of bone-resorbing OCs versus nonresorbing MGCs to generate a list of genes implicated in bone resorption. From this list of candidate genes, we investigated the role of Rho/Rnd3. Using primary RhoE-deficient OCs, we demonstrated that RhoE is indispensable for OC migration and bone resorption by maintaining fast actin turnover in podosomes. We further showed that RhoE activates podosome component cofilin by inhibiting its Rock-mediated phosphorylation. We conclude that the RhoE-Rock-cofilin pathway, by promoting podosome dynamics and patterning, is central for OC migration, SZ formation, and, ultimately, bone resorption.


Subject(s)
Actins/metabolism , Bone Resorption/metabolism , Osteoclasts/metabolism , rho GTP-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/metabolism , Amides/pharmacology , Animals , Bone Resorption/genetics , Cattle , Cell Differentiation/genetics , Cell Movement , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Giant Cells/metabolism , Humans , Mice , Mice, Transgenic , Phosphorylation , Pyridines/pharmacology , Transcriptome , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
13.
Genome Biol ; 14(7): R82, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23902802

ABSTRACT

BACKGROUND: The mouse inbred line C57BL/6J is widely used in mouse genetics and its genome has been incorporated into many genetic reference populations. More recently large initiatives such as the International Knockout Mouse Consortium (IKMC) are using the C57BL/6N mouse strain to generate null alleles for all mouse genes. Hence both strains are now widely used in mouse genetics studies. Here we perform a comprehensive genomic and phenotypic analysis of the two strains to identify differences that may influence their underlying genetic mechanisms. RESULTS: We undertake genome sequence comparisons of C57BL/6J and C57BL/6N to identify SNPs, indels and structural variants, with a focus on identifying all coding variants. We annotate 34 SNPs and 2 indels that distinguish C57BL/6J and C57BL/6N coding sequences, as well as 15 structural variants that overlap a gene. In parallel we assess the comparative phenotypes of the two inbred lines utilizing the EMPReSSslim phenotyping pipeline, a broad based assessment encompassing diverse biological systems. We perform additional secondary phenotyping assessments to explore other phenotype domains and to elaborate phenotype differences identified in the primary assessment. We uncover significant phenotypic differences between the two lines, replicated across multiple centers, in a number of physiological, biochemical and behavioral systems. CONCLUSIONS: Comparison of C57BL/6J and C57BL/6N demonstrates a range of phenotypic differences that have the potential to impact upon penetrance and expressivity of mutational effects in these strains. Moreover, the sequence variants we identify provide a set of candidate genes for the phenotypic differences observed between the two strains.


Subject(s)
Genome/genetics , Animals , Behavior, Animal , Disease Resistance/immunology , Eye/pathology , Female , Femur/diagnostic imaging , Hypersensitivity/immunology , INDEL Mutation/genetics , Killer Cells, Natural/immunology , Listeriosis/immunology , Listeriosis/microbiology , Male , Maze Learning , Mice, Inbred C57BL , Phenotype , Polymorphism, Single Nucleotide/genetics , Spleen/immunology , X-Ray Microtomography
14.
Glia ; 61(10): 1645-58, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23893349

ABSTRACT

As neuroinflammatory processes are involved in the pathogenesis of Parkinson's disease (PD), we provide several key data describing the time-course of microglial accumulation in relation with behavioral alterations and neurodegeneration in a murine model of PD induced by intrastriatal injection of 6-hydroxydopamine (6-OHDA). Our study argues for a major role of microglia which accumulation is somehow early and transient in spite of the neuronal loss progression. Moreover, we observed less 6-OHDA-induced neurodegeneration associated with less inflammatory reaction in DAP-12 Knock-In mice. The direct cell-to-cell contacts that may support physical interactions between microglia and altered dopaminergic neurons are ill-defined, while it is currently hypothesized that microglia support an immune-mediated amplification of neurodegeneration by establishing a molecular cross talk with neurons. Indeed, we sought to map microglia/neuron appositions in substantia nigra (SN) of 6-OHDA injected C57Bl/6 mice and CX3CR1/(GFP/+) mice. Confocal immunofluorescence analyses followed by 3D reconstitutions reveal close appositions between the soma of TH+ neurons and microglial cell bodies and ramifications. Interestingly, some microglial ramifications penetrated TH(+) somas and about 40% of GFP(+) microglial cells in the injured SN harbored TH(+) intracytoplasmic inclusions. These results suggest a direct cross talk between neurons and microglia that may exert a microphagocytic activity toward TH+ neurons. Altogether, these results obtained in a murine PD model may participate in the understanding of microglial cells' function in neurodegenerative diseases.


Subject(s)
Adrenergic Agents/toxicity , Cell Communication/physiology , Microglia/physiology , Neurons/physiology , Oxidopamine/toxicity , Parkinson Disease , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, Differentiation/metabolism , Apomorphine , Cell Communication/drug effects , Cell Communication/genetics , Cell Count , Disease Models, Animal , Dopamine Agonists , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Imaging, Three-Dimensional , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/cytology , Microglia/drug effects , Microscopy, Confocal , Neurons/cytology , Neurons/drug effects , Parkinson Disease/etiology , Parkinson Disease/genetics , Parkinson Disease/pathology , Receptors, Interleukin-8A/deficiency , Rotation , Substantia Nigra/pathology , Time Factors , Tyrosine 3-Monooxygenase/metabolism
15.
Int Rev Cell Mol Biol ; 305: 1-68, 2013.
Article in English | MEDLINE | ID: mdl-23890379

ABSTRACT

Mineralized tissues that are protective scaffolds in the most primitive species have evolved and acquired more specific functions in modern animals. These are as diverse as support in locomotion, ion homeostasis, and precise hormonal regulation. Bone formation is tightly controlled by a balance between anabolism, in which osteoblasts are the main players, and catabolism mediated by the osteoclasts. The bone matrix is deposited in a cyclic fashion during homeostasis and integrates several environmental cues. These include diffusible elements that would include estrogen or growth factors and physicochemical parameters such as bone matrix composition, stiffness, and mechanical stress. Therefore, the microenvironment is of paramount importance for controlling this delicate equilibrium. Here, we provide an overview of the most recent data highlighting the role of cell-adhesion molecules during bone formation. Due to the very large scope of the topic, we focus mainly on the role of the integrin receptor family during osteogenesis. Bone phenotypes of some deficient mice as well as diseases of human bones involving cell adhesion during this process are discussed in the context of bone physiology.


Subject(s)
Bone and Bones/metabolism , Osteogenesis , Signal Transduction , Animals , Bone and Bones/cytology , Cell Adhesion , Humans , Integrins/metabolism
16.
PLoS One ; 8(6): e65297, 2013.
Article in English | MEDLINE | ID: mdl-23776468

ABSTRACT

ITAM-bearing transmembrane signaling adaptors such as DAP12 and FcRγ are important players in bone homeostasis, but their precise role and functions are still unknown. It has been shown that osteoclast differentiation results from the integration of the RANK and of the DAP12 and FcRγ signaling pathways. DAP12-deficient mice suffer from a mild osteopetrosis and culture of their bone marrow cells in the presence of M-CSF and RANKL, fails to give rise to multinucleated osteoclasts. Here, we report that mice overexpressing human DAP12 have an osteopenic bone phenotype due to an increased number of osteoclasts on the surface of trabecular and cortical bone. This enhanced number of osteoclasts is associated with an increased number of proliferating myeloid progenitors in Tg-hDAP12 mice. It is concomitant with an arrest of B cell development at the Pre-Pro B/Pre B stage in the bone marrow of Tg-hDAP12 mice and important decrease of follicular and marginal B cells in the spleen of these animals. Our data show that the overexpression of DAP12 results in both increased osteoclastogenesis and impaired hematopoiesis underlining the relationship between bone homeostasis and hematopoiesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Diseases, Metabolic/metabolism , Gene Expression Regulation , Hematopoiesis/physiology , Membrane Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Diseases, Metabolic/genetics , Cell Proliferation , Cells, Cultured , Female , Flow Cytometry , Hematopoiesis/genetics , Humans , Membrane Proteins/genetics , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology
17.
FASEB J ; 27(9): 3608-18, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23742809

ABSTRACT

In osteoclasts, Src controls podosome organization and bone degradation, which leads to an osteopetrotic phenotype in src(-/-) mice. Since this phenotype was even more severe in src(-/-)hck(-/-) mice, we examined the individual contribution of Hck in bone homeostasis. Compared to wt mice, hck(-/-) mice exhibited an osteopetrotic phenotype characterized by an increased density of trabecular bone and decreased bone degradation, although osteoclastogenesis was not impaired. Podosome organization and matrix degradation were found to be defective in hck(-/-) osteoclast precursors (preosteoclast) but were normal in mature hck(-/-) osteoclasts, probably through compensation by Src, which was specifically overexpressed in mature osteoclasts. As a consequence of podosome defects, the 3-dimensional migration of hck(-/-) preosteoclasts was strongly affected in vitro. In vivo, this translated by altered bone homing of preosteoclasts in hck(-/-) mice: in metatarsals of 1-wk-old mice, when bone formation strongly depends on the recruitment of these cells, reduced numbers of osteoclasts and abnormal developing trabecular bone were observed. This phenotype was still detectable in adults. In summmary, Hck is one of the very few effectors of preosteoclast recruitment described to date and thereby plays a critical role in bone remodeling.


Subject(s)
Bone and Bones/cytology , Bone and Bones/metabolism , Cell Movement/physiology , Osteoclasts/cytology , Osteopetrosis/metabolism , Proto-Oncogene Proteins c-hck/metabolism , Animals , Cell Movement/genetics , Cells, Cultured , Female , Homeostasis/genetics , Homeostasis/physiology , Male , Mice , Mice, Knockout , Osteoclasts/metabolism , Osteopetrosis/genetics , Proto-Oncogene Proteins c-hck/genetics , src-Family Kinases/genetics , src-Family Kinases/metabolism
18.
PLoS One ; 8(2): e56865, 2013.
Article in English | MEDLINE | ID: mdl-23441221

ABSTRACT

Dendritic cells initiate adaptive immune responses, leading either to control cancer by effector T cells or to exacerbate cancer by regulatory T cells that inhibit IFN-γ-mediated Th1-type response. Dendritic cells can also induce Th17-type immunity, mediated by IL-17A. However, the controversial role of this cytokine in cancer requires further investigations. We generated dendritic cells from peripheral blood monocytes to investigate lifespan, phenotype and chemoresistance of dendritic cells, treated with IL-17A with or without IFN-γ. Studying the expression of Bcl-2 family members, we demonstrated that dendritic cells constitutively express one pro-survival Bcl-2 member: MCL1. Immature dendritic cells were CD40(low)HLADR(low) CD1a(+) MCL1(+), did not express CD14, CD68 or BCL2A1, and displayed a short 2-day lifespan. IL-17A-treated DC exhibited a semi-mature (CD40(high) HLADR(low)) pre-M2 (CCL22(+) CD206(+) CD163(+) IL1RN(+) IL-10(-) CXCL10(-) IL-12(-)) mixed (CD1a(+) CD14+ CD68(+)) macrophage-dendritic cell phenotype. They efficiently exerted mannose receptor-mediated endocytosis and did not produce superoxide anions, in the absence of TLR engagement. Interestingly, IL-17A promoted a long-term survival of dendritic cells, beyond 12 days, that correlated to BCL2A1 induction, a pro-survival Bcl-2 family member. BCL2A1 transcription was activated by NF-κB, downstream of IL-17A transduction. Thus, immature dendritic cells only express MCL1, whereas IL-17A-treated dendritic cells concomitantly expressed two pro-survival Bcl-2 family members: MCL1 and BCL2A1. These latter developed chemoresistance to 11 of the 17 chemotherapy agents tested. However, high doses of either vinblastine or cytarabine decreased MCL1 expression and induced dendritic cell death. When IL-17A is produced in vivo, administration of anti-IL-17A biotherapy may impair dendritic cell survival by targeting BCL2A1 expression. Consequently, depending on the effector or regulatory role of dendritic cells, blocking IL-17A, may be either dangerous or beneficial for cancer outcomes, thus contributing to the apparent controversy around the role of IL-17A in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Drug Resistance, Neoplasm , Interleukin-17/pharmacology , Monocytes/cytology , Cell Differentiation , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dendritic Cells/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation/drug effects , Humans , Immunophenotyping , Interferon-gamma/pharmacology , Minor Histocompatibility Antigens , Myeloid Cell Leukemia Sequence 1 Protein , NF-kappa B/metabolism , Phenotype , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription, Genetic
19.
J Cell Sci ; 125(Pt 16): 3790-800, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22553210

ABSTRACT

Multinucleated muscle fibres arise by fusion of precursor cells called myoblasts. We previously showed that CKIP-1 ectopic expression in C2C12 myoblasts increased cell fusion. In this work, we report that CKIP-1 depletion drastically impairs C2C12 myoblast fusion in vitro and in vivo during zebrafish muscle development. Within developing fast-twich myotome, Ckip-1 localises at the periphery of fast precursor cells, closed to the plasma membrane. Unlike wild-type myoblasts that form spatially arrayed multinucleated fast myofibres, Ckip-1-deficient myoblasts show a drastic reduction in fusion capacity. A search for CKIP-1 binding partners identified the ARPC1 subunit of Arp2/3 actin nucleation complex essential for myoblast fusion. We demonstrate that CKIP-1, through binding to plasma membrane phosphoinositides via its PH domain, regulates cell morphology and lamellipodia formation by recruiting the Arp2/3 complex at the plasma membrane. These results establish CKIP-1 as a regulator of cortical actin that recruits the Arp2/3 complex at the plasma membrane essential for muscle precursor elongation and fusion.


Subject(s)
Carrier Proteins/physiology , Membrane Fusion/physiology , Myoblasts/cytology , Actin-Related Protein 2-3 Complex/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Communication/physiology , Cell Differentiation/physiology , Cell Fusion , Cell Line , Humans , Intracellular Signaling Peptides and Proteins , Mammals , Mice , Myoblasts/metabolism , Transfection , Zebrafish
20.
J Clin Invest ; 122(5): 1791-802, 2012 May.
Article in English | MEDLINE | ID: mdl-22505457

ABSTRACT

Autoimmunity is complicated by bone loss. In human rheumatoid arthritis (RA), the most severe inflammatory joint disease, autoantibodies against citrullinated proteins are among the strongest risk factors for bone destruction. We therefore hypothesized that these autoantibodies directly influence bone metabolism. Here, we found a strong and specific association between autoantibodies against citrullinated proteins and serum markers for osteoclast-mediated bone resorption in RA patients. Moreover, human osteoclasts expressed enzymes eliciting protein citrullination, and specific N-terminal citrullination of vimentin was induced during osteoclast differentiation. Affinity-purified human autoantibodies against mutated citrullinated vimentin (MCV) not only bound to osteoclast surfaces, but also led to robust induction of osteoclastogenesis and bone-resorptive activity. Adoptive transfer of purified human MCV autoantibodies into mice induced osteopenia and increased osteoclastogenesis. This effect was based on the inducible release of TNF-α from osteoclast precursors and the subsequent increase of osteoclast precursor cell numbers with enhanced expression of activation and growth factor receptors. Our data thus suggest that autoantibody formation in response to citrullinated vimentin directly induces bone loss, providing a link between the adaptive immune system and bone.


Subject(s)
Autoantibodies/metabolism , Bone Resorption/immunology , Citrulline/immunology , Osteoclasts/physiology , Vimentin/immunology , Animals , Antibody Specificity , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , Autoantibodies/blood , Autoantibodies/isolation & purification , Biomarkers/blood , Bone and Bones/immunology , Bone and Bones/pathology , Case-Control Studies , Cell Differentiation , Cells, Cultured , Collagen Type I/blood , Humans , Hydrolases/metabolism , Mice , Mice, Transgenic , Osteoclasts/enzymology , Osteoclasts/metabolism , Protein Processing, Post-Translational , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases , Tumor Necrosis Factor-alpha/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...