Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Cell Biochem ; 121(4): 2927-2937, 2020 04.
Article in English | MEDLINE | ID: mdl-31692073

ABSTRACT

Bone resorption and organelle homeostasis in osteoclasts require specialized intracellular trafficking. Sorting nexin 10 (Snx10) is a member of the sorting nexin family of proteins that plays crucial roles in cargo sorting in the endosomal pathway by its binding to phosphoinositide(3)phosphate (PI3P) localized in early endosomes. We and others have shown previously that the gene encoding sorting Snx10 is required for osteoclast morphogenesis and function, as osteoclasts from humans and mice lacking functional Snx10 are dysfunctional. To better understand the role and mechanisms by which Snx10 regulates vesicular transport, the aim of the present work was to study PIKfyve, another PI3P-binding protein, which phosphorylates PI3P to PI(3,5)P2. PI(3,5)P2 is known to be required for endosome/lysosome maturation, and the inhibition of PIKfyve causes endosome enlargement. Overexpression of Snx10 also induces accumulation of early endosomes suggesting that both Snx10 and PIKfyve are required for normal endosome/lysosome transition. Apilimod is a small molecule with specific, nanomolar inhibitory activity on PIKfyve but only in the presence of key osteoclast factors CLCN7, OSTM1, and Snx10. This observation suggests that apilimod's inhibitory effects are mediated by endosome/lysosome disruption. Here we show that both Snx10 and PIKfyve colocalize to early endosomes in osteoclasts and coimmunoprecipitate in vesicle fractions. Treatment with 10 nM apilimod or genetic deletion of PIKfyve in cells resulted in the accumulation of early endosomes, and in the inhibition of osteoclast differentiation, lysosome formation, and secretion of TRAP from differentiated osteoclasts. Snx10 and PIKfyve also colocalized in gastric zymogenic cells, another cell type impacted by Snx10 mutations. Apilimod-specific inhibition of PIKfyve required Snx10 expression, as it did not inhibit lysosome biogenesis in Snx10-deficient osteoclasts. These findings suggest that Snx10 and PIKfyve are involved in the regulation of endosome/lysosome homeostasis via the synthesis of PI(3,5)P2 and may point to a new strategy to prevent bone loss.


Subject(s)
Lysosomes/metabolism , Osteoclasts/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Sorting Nexins/metabolism , Animals , Biological Transport , Bone Resorption/metabolism , Cytoplasm/metabolism , Endosomes/metabolism , Gene Expression Regulation , Hydrazones/pharmacology , Mice , Morpholines/pharmacology , Phosphorylation , Pyrimidines/pharmacology , RAW 264.7 Cells
2.
ACS Chem Biol ; 12(10): 2619-2630, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28849908

ABSTRACT

Histone acetyltransferases of the MYST family are recruited to chromatin by BRPF scaffolding proteins. We explored functional consequences and the therapeutic potential of inhibitors targeting acetyl-lysine dependent protein interaction domains (bromodomains) present in BRPF1-3 in bone maintenance. We report three potent and selective inhibitors: one (PFI-4) with high selectivity for the BRPF1B isoform and two pan-BRPF bromodomain inhibitors (OF-1, NI-57). The developed inhibitors displaced BRPF bromodomains from chromatin and did not inhibit cell growth and proliferation. Intriguingly, the inhibitors impaired RANKL-induced differentiation of primary murine bone marrow cells and human primary monocytes into bone resorbing osteoclasts by specifically repressing transcriptional programs required for osteoclastogenesis. The data suggest a key role of BRPF in regulating gene expression during osteoclastogenesis, and the excellent druggability of these bromodomains may lead to new treatment strategies for patients suffering from bone loss or osteolytic malignant bone lesions.


Subject(s)
Bone Marrow Cells/physiology , Carrier Proteins/metabolism , Cell Differentiation/physiology , Osteoclasts/physiology , Animals , Carrier Proteins/genetics , Computational Biology , Humans , Models, Molecular , Multigene Family , Protein Array Analysis , Protein Conformation , Protein Domains , Stem Cells
3.
Connect Tissue Res ; 57(3): 161-74, 2016 May.
Article in English | MEDLINE | ID: mdl-26818783

ABSTRACT

Three named cell types degrade and remove skeletal tissues during growth, repair, or disease: osteoclasts, chondroclasts, and septoclasts. A fourth type, unnamed and less understood, removes nonmineralized cartilage during development of secondary ossification centers. "Osteoclasts," best known and studied, are polykaryons formed by fusion of monocyte precursors under the influence of colony stimulating factor 1 (CSF)-1 (M-CSF) and RANKL. They resorb bone during growth, remodeling, repair, and disease. "Chondroclasts," originally described as highly similar in cytological detail to osteoclasts, reside on and degrade mineralized cartilage. They may be identical to osteoclasts since to date there are no distinguishing markers for them. Because osteoclasts also consume cartilage cores along with bone during growth, the term "chondroclast" might best be reserved for cells attached only to cartilage. "Septoclasts" are less studied and appreciated. They are mononuclear perivascular cells rich in cathepsin B. They extend a cytoplasmic projection with a ruffled membrane and degrade the last transverse septum of hypertrophic cartilage in the growth plate, permitting capillaries to bud into it. To do this, antiangiogenic signals in cartilage must give way to vascular trophic factors, mainly vascular endothelial growth factor (VEGF). The final cell type excavates cartilage canals for vascular invasion of articular cartilage during development of secondary ossification centers. The "clasts" are considered in the context of fracture repair and diseases such as arthritis and tumor metastasis. Many observations support an essential role for hypertrophic chondrocytes in recruiting septoclasts and osteoclasts/chondroclasts by supplying VEGF and RANKL. The intimate relationship between blood vessels and skeletal turnover and repair is also examined.


Subject(s)
Bone Development , Chondrocytes/pathology , Disease , Neovascularization, Pathologic/pathology , Osteoclasts/pathology , Wound Healing , Animals , Humans
4.
PLoS One ; 10(6): e0128275, 2015.
Article in English | MEDLINE | ID: mdl-26042409

ABSTRACT

The fusion of monocyte/macrophage lineage cells into fully active, multinucleated, bone resorbing osteoclasts is a complex cell biological phenomenon that utilizes specialized proteins. OC-STAMP, a multi-pass transmembrane protein, has been shown to be required for pre-osteoclast fusion and for optimal bone resorption activity. A previously reported knockout mouse model had only mononuclear osteoclasts with markedly reduced resorption activity in vitro, but with paradoxically normal skeletal micro-CT parameters. To further explore this and related questions, we used mouse ES cells carrying a gene trap allele to generate a second OC-STAMP null mouse strain. Bone histology showed overall normal bone form with large numbers of TRAP-positive, mononuclear osteoclasts. Micro-CT parameters were not significantly different between knockout and wild type mice at 2 or 6 weeks old. At 6 weeks, metaphyseal TRAP-positive areas were lower and mean size of the areas were smaller in knockout femora, but bone turnover markers in serum were normal. Bone marrow mononuclear cells became TRAP-positive when cultured with CSF-1 and RANKL, but they did not fuse. Expression levels of other osteoclast markers, such as cathepsin K, carbonic anhydrase II, and NFATc1, were not significantly different compared to wild type. Actin rings were present, but small, and pit assays showed a 3.5-fold decrease in area resorbed. Restoring OC-STAMP in knockout cells by lentiviral transduction rescued fusion and resorption. N- and C-termini of OC-STAMP were intracellular, and a predicted glycosylation site was shown to be utilized and to lie on an extracellular loop. The site is conserved in all terrestrial vertebrates and appears to be required for protein stability, but not for fusion. Based on this and other results, we present a topological model of OC-STAMP as a 6-transmembrane domain protein. We also contrast the osteoclast-specific roles of OC- and DC-STAMP with more generalized cell fusion mechanisms.


Subject(s)
Cell Fusion , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Acid Phosphatase/metabolism , Alleles , Amino Acid Motifs , Amino Acid Sequence , Animals , Biomarkers/metabolism , Bone Resorption/pathology , Cell Survival , Conserved Sequence , Femur/metabolism , Femur/pathology , Gene Expression Regulation , Glycosylation , HEK293 Cells , Humans , Isoenzymes/metabolism , Lentivirus/metabolism , Membrane Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Molecular Sequence Data , Osteoclasts/enzymology , Osteogenesis , Tartrate-Resistant Acid Phosphatase , Transduction, Genetic
5.
PLoS One ; 10(5): e0127537, 2015.
Article in English | MEDLINE | ID: mdl-25992615

ABSTRACT

Plekhm1 is a large, multi-modular, adapter protein implicated in osteoclast vesicle trafficking and bone resorption. In patients, inactivating mutations cause osteopetrosis, and gain-of-function mutations cause osteopenia. Investigations of potential Plekhm1 interaction partners by mass spectrometry identified TRAFD1 (FLN29), a protein previously shown to suppress toll-like receptor signaling in monocytes/macrophages, thereby dampening inflammatory responses to innate immunity. We mapped the binding domains to the TRAFD1 zinc finger (aa 37-60), and to the region of Plekhm1 between its second pleckstrin homology domain and its C1 domain (aa 784-986). RANKL slightly increased TRAFD1 levels, particularly in primary osteoclasts, and the co-localization of TRAFD1 with Plekhm1 also increased with RANKL treatment. Stable knockdown of TRAFD1 in RAW 264.7 cells inhibited resorption activity proportionally to the degree of knockdown, and inhibited acidification. The lack of acidification occurred despite the presence of osteoclast acidification factors including carbonic anhydrase II, a3-V-ATPase, and the ClC7 chloride channel. Secretion of TRAP and cathepsin K were also markedly inhibited in knockdown cells. Truncated Plekhm1 in ia/ia osteopetrotic rat cells prevented vesicle localization of Plekhm1 and TRAFD1. We conclude that TRAFD1, in association with Plekhm1/Rab7-positive late endosomes-early lysosomes, has a previously unknown role in vesicle trafficking, acidification, and resorption in osteoclasts.


Subject(s)
Acids/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Glycoproteins/metabolism , Osteoclasts/metabolism , Animals , Autophagy-Related Proteins , Cell Differentiation , Intracellular Signaling Peptides and Proteins/genetics , Mice , Osteoclasts/cytology , Protein Binding , RNA, Messenger/genetics , Rats
6.
PLoS Genet ; 11(3): e1005057, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25811986

ABSTRACT

Mutations in sorting nexin 10 (Snx10) have recently been found to account for roughly 4% of all human malignant osteopetrosis, some of them fatal. To study the disease pathogenesis, we investigated the expression of Snx10 and created mouse models in which Snx10 was knocked down globally or knocked out in osteoclasts. Endocytosis is severely defective in Snx10-deficient osteoclasts, as is extracellular acidification, ruffled border formation, and bone resorption. We also discovered that Snx10 is highly expressed in stomach epithelium, with mutations leading to high stomach pH and low calcium solubilization. Global Snx10-deficiency in mice results in a combined phenotype: osteopetrosis (due to osteoclast defect) and rickets (due to high stomach pH and low calcium availability, resulting in impaired bone mineralization). Osteopetrorickets, the paradoxical association of insufficient mineralization in the context of a positive total body calcium balance, is thought to occur due to the inability of the osteoclasts to maintain normal calcium-phosphorus homeostasis. However, osteoclast-specific Snx10 knockout had no effect on calcium balance, and therefore led to severe osteopetrosis without rickets. Moreover, supplementation with calcium gluconate rescued mice from the rachitic phenotype and dramatically extended life span in global Snx10-deficient mice, suggesting that this may be a life-saving component of the clinical approach to Snx10-dependent human osteopetrosis that has previously gone unrecognized. We conclude that tissue-specific effects of Snx10 mutation need to be considered in clinical approaches to this disease entity. Reliance solely on hematopoietic stem cell transplantation can leave hypocalcemia uncorrected with sometimes fatal consequences. These studies established an essential role for Snx10 in bone homeostasis and underscore the importance of gastric acidification in calcium uptake.


Subject(s)
Bone Density/genetics , Gastric Acid/metabolism , Osteoclasts/metabolism , Osteopetrosis/genetics , Sorting Nexins/genetics , Amino Acid Sequence , Animals , Calcium/administration & dosage , Calcium/metabolism , Calcium Gluconate/administration & dosage , Endocytosis/genetics , Gene Knockdown Techniques , Homeostasis , Humans , Mice , Mutation , Osteoclasts/drug effects , Osteoclasts/pathology , Osteopetrosis/metabolism , Osteopetrosis/pathology , Sorting Nexins/metabolism , Vitamin D Deficiency/genetics , Vitamin D Deficiency/metabolism , Vitamin D Deficiency/pathology
7.
Cell Host Microbe ; 17(1): 58-71, 2015 Jan 14.
Article in English | MEDLINE | ID: mdl-25500191

ABSTRACT

The host endolysosomal compartment is often manipulated by intracellular bacterial pathogens. Salmonella (Salmonella enterica serovar Typhimurium) secrete numerous effector proteins, including SifA, through a specialized type III secretion system to hijack the host endosomal system and generate the Salmonella-containing vacuole (SCV). To form this replicative niche, Salmonella targets the Rab7 GTPase to recruit host membranes through largely unknown mechanisms. We show that Pleckstrin homology domain-containing protein family member 1 (PLEKHM1), a lysosomal adaptor, is targeted by Salmonella through direct interaction with SifA. By binding the PLEKHM1 PH2 domain, Salmonella utilize a complex containing PLEKHM1, Rab7, and the HOPS tethering complex to mobilize phagolysosomal membranes to the SCV. Depletion of PLEKHM1 causes a profound defect in SCV morphology with multiple bacteria accumulating in enlarged structures and significantly dampens Salmonella proliferation in multiple cell types and mice. Thus, PLEKHM1 provides a critical interface between pathogenic infection and the host endolysosomal system.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bacterial Proteins/metabolism , Glycoproteins/metabolism , Host-Pathogen Interactions , Membrane Glycoproteins/metabolism , Salmonella typhimurium/growth & development , Vacuoles/microbiology , Animals , Autophagy-Related Proteins , Carrier Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/metabolism , Protein Binding , Protein Interaction Mapping , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
8.
J Orthop Res ; 32(12): 1562-71, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25125336

ABSTRACT

Because Chondrosarcoma is resistant to available chemotherapy and radiation regimens, wide resection is the mainstay in treatment, which frequently results in high morbidity and which may not prevent local recurrence. There is a clear need for improved adjuvant treatment of this malignancy. We have observed the presence of osteoclasts in the microenvironment of chondrosarcoma in human pathological specimens. We utilized the Swarm rat chondrosarcoma (SRC) model to test the hypothesis that osteoclasts affect chondrosarcoma pathogenesis. We implanted SRC tumors in tibia of Sprague-Dawley rats and analyzed bone histologically and radiographically for bone destruction and tumor growth. At three weeks, tumors invaded local bone causing cortical disruption and trabecular resorption. Bone destruction was accompanied by increased osteoclast number and resorbed bone surface. Treatment of rats with the zoledronic acid prevented cortical destruction, inhibited trabecular resorption, and resulted in decreased tumor volume in bone. To confirm that inhibition of osteoclasts per se, and not off-target effects of drug, was responsible for the prevention of tumor growth and bone destruction, we implanted SRC into osteopetrotic rat tibia. SRC-induced bone destruction and tumor growth were impaired in osteopetrotic bone compared with control bone. The results from our animal model demonstrate that osteoclasts contribute to chondrosarcoma-mediated bone destruction and tumor growth and may represent a therapeutic target in particular chondrosarcoma patients.


Subject(s)
Bone and Bones/pathology , Chondrosarcoma/pathology , Osteoclasts/physiology , Animals , Bone Resorption/etiology , Bone Resorption/prevention & control , Diphosphonates/pharmacology , Imidazoles/pharmacology , Male , Parathyroid Hormone-Related Protein/analysis , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta2/analysis , Zoledronic Acid
9.
J Cell Biol ; 204(7): 1083-6, 2014 Mar 31.
Article in English | MEDLINE | ID: mdl-24687277

ABSTRACT

The mitochondrial inner membrane contains a large protein complex that functions in inner membrane organization and formation of membrane contact sites. The complex was variably named the mitochondrial contact site complex, mitochondrial inner membrane organizing system, mitochondrial organizing structure, or Mitofilin/Fcj1 complex. To facilitate future studies, we propose to unify the nomenclature and term the complex "mitochondrial contact site and cristae organizing system" and its subunits Mic10 to Mic60.


Subject(s)
Mitochondrial Membranes/chemistry , Mitochondrial Proteins/chemistry , Protein Subunits/chemistry , Animals , Humans , Models, Molecular , Multiprotein Complexes/chemistry , Terminology as Topic
10.
PLoS One ; 5(4): e9959, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20376364

ABSTRACT

BACKGROUND: Investigations of naturally-occurring mutations in animal models provide important insights and valuable disease models. Lamins A and C, along with lamin B, are type V intermediate filament proteins which constitute the proteinaceous boundary of the nucleus. LMNA mutations in humans cause a wide range of phenotypes, collectively termed laminopathies. To identify the mutation and investigate the phenotype of a spontaneous, semi-dominant mutation that we have named Disheveled hair and ear (Dhe), which causes a sparse coat and small external ears in heterozygotes and lethality in homozygotes by postnatal day 10. FINDINGS: Genetic mapping identified a point mutation in the Lmna gene, causing a single amino acid change, L52R, in the coiled coil rod domain of lamin A and C proteins. Cranial sutures in Dhe/+ mice failed to close. Gene expression for collagen types I and III in sutures was deficient. Skulls were small and disproportionate. Skeletons of Dhe/+ mice were hypomineralized and total body fat was deficient in males. In homozygotes, skin and oral mucosae were dysplastic and ulcerated. Nuclear morphometry of cultured cells revealed gene dose-dependent blebbing and wrinkling. CONCLUSION: Dhe mice should provide a useful new model for investigations of the pathogenesis of laminopathies.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Disease Models, Animal , Lamin Type A/genetics , Phosphoproteins/genetics , Point Mutation , Animals , Dishevelled Proteins , Female , Genotype , Humans , Male , Mice , Mice, Knockout , Phenotype
11.
Biochem Mol Biol Educ ; 38(6): 385-92, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21567867

ABSTRACT

A key goal of molecular/cell biology/biotechnology is to identify essential genes in virtually every physiological process to uncover basic mechanisms of cell function and to establish potential targets of drug therapy combating human disease. This article describes a semester-long, project-oriented molecular/cellular/biotechnology laboratory providing students, within a framework of bone cell biology, with a modern approach to gene discovery. Students are introduced to the topics of bone cells, bone synthesis, bone resorption, and osteoporosis. They then review the theory of microchip gene arrays, and study microchip array data generated during the differentiation of bone-resorbing osteoclasts in vitro. The class selects genes whose expression increases during osteoclastogenesis, and researches them in small groups using web-based bioinformatics tools. Students then go to a biotechnology company website to find and order small inhibitory RNAs (siRNAs) designed to "knockdown" expression of the gene of interest. Students then learn to transfect these siRNAs into osteoclasts, stimulate the cells to differentiate, assay osteoclast differentiation in vitro, and measure specific gene expression using real-time PCR and immunoblotting. Specific siRNA knockdown resulting in a decrease in osteoclastogenesis is indicative of a gene's physiological relevance. The results are analyzed statistically and presented to the class in groups. In the past 2 years, students identified several genes essential for optimal osteoclast differentiation, including Myo1d. The students hypothesize that the myo1d protein functions in osteoclasts to deliver important proteins to the cell surface via vesicular transport along microfilaments. Student response to the new course was overwhelmingly positive.

12.
Antioxid Redox Signal ; 13(1): 27-37, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-19951071

ABSTRACT

The central role of reactive oxygen species (ROS) in osteoclast differentiation and in bone homeostasis prompted us to characterize the redox regulatory system of osteoclasts. In this report, we describe the expression and functional characterization of PAMM, a CXXC motif-containing peroxiredoxin 2-like protein expressed in bone marrow monocytes on stimulation with M-CSF and RANKL. Expression of wild-type (but not C to G mutants of the CXXC domain) PAMM in HEK293 cells results in an increased GSH/GSSG ratio, indicating a shift toward a more reduced environment. Expression of PAMM in RAW264.7 monocytes protected cells from hydrogen peroxide-induced oxidative stress, indicating that PAMM regulates cellular redox status. RANKL stimulation of RAW 264.7 cells caused a decrease in the GSH/GSSG ratio (reflecting a complementary increase in ROS). In addition, RANKL-induced osteoclast formation requires phosphorylation and translocation of NF-kappaB and c-Jun. In stably transfected RAW 264.7 cells, PAMM overexpression prevented the reduction of GSH/GSSG induced by RANKL. Concurrently, PAMM expression completely abolished RANKL-induced p100 NF-kappaB and c-Jun activation, as well as osteoclast formation. We conclude that PAMM is a redox regulatory protein that modulates osteoclast differentiation in vitro. PAMM expression may affect bone resorption in vivo and help to maintain bone mass.


Subject(s)
Cell Differentiation , Osteoclasts/cytology , Osteoclasts/metabolism , Peroxiredoxins/metabolism , Amino Acid Sequence , Animals , Cell Survival , Cells, Cultured , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Molecular Sequence Data , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Oxidative Stress , Peroxiredoxins/chemistry , Peroxiredoxins/genetics , RNA, Messenger/genetics , Rats , Rats, Mutant Strains , Reverse Transcriptase Polymerase Chain Reaction
13.
Am J Pathol ; 175(6): 2668-75, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19893052

ABSTRACT

The septoclast is a specialized, cathepsin B-rich, perivascular cell type that accompanies invading capillaries on the metaphyseal side of the growth plate during endochondral bone growth. The putative role of septoclasts is to break down the terminal transverse septum of growth plate cartilage and permit capillaries to bud into the lower hypertrophic zone. This process fails in osteoclast-deficient, osteopetrotic animal models, resulting in a progressive growth plate dysplasia. The toothless rat is severely osteopetrotic because of a frameshift mutation in the colony-stimulating factor-1 (CSF-1) gene (Csf1(tl)). Whereas CSF-1 injections quickly restore endosteal osteoclast populations, they do not improve the chondrodysplasia. We therefore investigated septoclast populations in Csf1(tl)/Csf1(tl) rats and wild-type littermates, with and without CSF-1 treatment, at 2 weeks, before the dysplasia is pronounced, and at 4 weeks, by which time it is severe. Tibial sections were immunolabeled for cathepsin B and septoclasts were counted. Csf1(tl)/Csf1(tl) mutants had significant reductions in septoclasts at both times, although they were more pronounced at 4 weeks. CSF-1 injections increased counts in wild-type and mutant animals at both times, restoring mutants to normal levels at 2 weeks. In all of the mutants, septoclasts seemed misoriented and had abnormal ultrastructure. We conclude that CSF-1 promotes angiogenesis at the chondroosseous junction, but that, in Csf1(tl)/Csf1(tl) rats, septoclasts are unable to direct their degradative activity appropriately, implying a capillary guidance role for locally supplied CSF-1.


Subject(s)
Bone Development/physiology , Chondrocytes/pathology , Growth Plate/pathology , Macrophage Colony-Stimulating Factor/deficiency , Neovascularization, Physiologic/physiology , Osteopetrosis/metabolism , Animals , Bone Diseases, Developmental/drug therapy , Bone Diseases, Developmental/genetics , Bone Diseases, Developmental/metabolism , Cartilage/blood supply , Cartilage/cytology , Growth Plate/drug effects , Growth Plate/metabolism , Immunohistochemistry , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/pharmacology , Osteopetrosis/genetics , Rats , Rats, Mutant Strains
14.
J Cell Physiol ; 215(2): 497-505, 2008 May.
Article in English | MEDLINE | ID: mdl-18064667

ABSTRACT

Microarray and real-time RT-PCR were used to examine expression changes in primary bone marrow cells and RAW 264.7 cells in response to RANKL. In silico sequence analysis was performed on a novel gene which we designate OC-STAMP. Specific siRNA and antibodies were used to inhibit OC-STAMP RNA and protein, respectively, and tartrate-resistant acid phosphatase (TRAP)+ multinucleated osteoclasts were counted. Antibodies were used to probe bone tissues and western blots of RAW cell extracts +/- RANKL. cDNA overexpression constructs were transfected into RAW cells and the effect on RANKL-induced differentiation was studied. OC-STAMP was very strongly up-regulated during osteoclast differentiation. Northern blots and sequence analysis revealed two transcripts of 2 and 3.7 kb differing only in 3'UTR length, consistent with predictions from genome sequence. The mRNA encodes a 498 amino acid, multipass transmembrane protein that is highly conserved in mammals. It has little overall homology to other proteins. The carboxy-terminal 193 amino acids, however, are significantly similar to the DC-STAMP family consensus sequence. DC-STAMP is a transmembrane protein required for osteoclast precursor fusion. Knockdown of OC-STAMP mRNA by siRNA and protein inhibition by antibodies significantly suppressed the formation of TRAP+, multinucleated cells in differentiating osteoclast cultures, with many TRAP+ mononuclear cells present. Conversely, overexpression of OC-STAMP increased osteoclastic differentiation of RAW 264.7 cells. We conclude that OC-STAMP is a previously unknown, RANKL-induced, multipass transmembrane protein that promotes the formation of multinucleated osteoclasts.


Subject(s)
Cell Differentiation/physiology , Membrane Proteins/physiology , Osteoclasts/cytology , Osteoclasts/metabolism , RANK Ligand/pharmacology , Acid Phosphatase/metabolism , Amino Acid Sequence , Animals , Antibodies/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone and Bones/metabolism , Cell Differentiation/drug effects , Cell Nucleus/ultrastructure , Cells, Cultured , Immunohistochemistry , Isoenzymes/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Microarray Analysis , Molecular Sequence Data , Osteoclasts/drug effects , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis , Tartrate-Resistant Acid Phosphatase , Transfection , Up-Regulation
15.
J Cell Physiol ; 214(1): 56-64, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17541940

ABSTRACT

Bone morphogenetic proteins (BMPs) play pivotal roles in bone and cartilage growth and repair. Through phenotypes of short-ear (se) mice, which have BMP-5 mutations, a role for BMP-5 in some specific aspects of skeletogenesis and cartilage growth is known. This report examines BMP-5 expression in the growth plate and in differentiating cultures of primary chondrocytes, and the effects of addition of BMP-5 or its inhibition by anti-BMP-5 antibody in chondrocyte cultures. By laser capture microdissection and immunohistochemistry, we found that BMP-5 is expressed in proliferating zone (PZ) chondrocytes and that the expression increases sharply with hypertrophic differentiation. A similar pattern was observed in differentiating cultures of primary chondrocytes, with BMP-5 expression increasing as cells differentiated, in contrast to other BMPs. BMP-5 added to cultures increased cell proliferation early in the culture period and also stimulated cartilage matrix synthesis. Also, BMP-5 addition to the cultures activated phosphorylation of Smad 1/5/8 and p38 MAP kinase and caused increased nuclear accumulation of phospho-Smads. Anti-BMP-5 antibody inhibited the endogenous BMP-5, reducing cell proliferation and phospho-Smad nuclear accumulation. Together, the results demonstrate that BMP-5 is normally an important regulator of chondrocyte proliferation and differentiation. Whether other BMPs may compensate in BMP-5 loss-of-function mutations is discussed.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , Cell Proliferation , Chondrocytes/metabolism , Extracellular Matrix/metabolism , Animals , Bone Morphogenetic Protein 5 , Calcification, Physiologic/physiology , Cartilage, Articular/cytology , Cartilage, Articular/enzymology , Cells, Cultured , Chondrocytes/cytology , Glycosaminoglycans/analysis , Immunohistochemistry , In Vitro Techniques , Rats , Ribs/cytology
16.
J Clin Invest ; 117(4): 919-30, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17404618

ABSTRACT

This study illustrates that Plekhm1 is an essential protein for bone resorption, as loss-of-function mutations were found to underlie the osteopetrotic phenotype of the incisors absent rat as well as an intermediate type of human osteopetrosis. Electron and confocal microscopic analysis demonstrated that monocytes from a patient homozygous for the mutation differentiated into osteoclasts normally, but when cultured on dentine discs, the osteoclasts failed to form ruffled borders and showed little evidence of bone resorption. The presence of both RUN and pleckstrin homology domains suggests that Plekhm1 may be linked to small GTPase signaling. We found that Plekhm1 colocalized with Rab7 to late endosomal/lysosomal vesicles in HEK293 and osteoclast-like cells, an effect that was dependent on the prenylation of Rab7. In conclusion, we believe PLEKHM1 to be a novel gene implicated in the development of osteopetrosis, with a putative critical function in vesicular transport in the osteoclast.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Chromosomes, Human, Pair 10 , Membrane Glycoproteins/genetics , Osteopetrosis/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Autophagy-Related Proteins , Chromosome Mapping , Female , Gene Expression Regulation , Humans , Kidney/physiology , Kidney/physiopathology , Male , Membrane Glycoproteins/metabolism , Monocytes/physiology , Mutation , Organ Specificity , Pedigree , Rats , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
17.
J Cell Biochem ; 101(2): 399-410, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17171642

ABSTRACT

The success of vascular intervention including angioplasty, stenting, and arterial bypass remains limited by negative remodeling resulted in lumen restenosis. This study was to characterize the global transcription profile reflecting concurrent events along arterial remodeling and neointima formation in a rat carotid artery balloon-injury model. Expression profiling of injured and control common carotid arteries on days 4, 7, 14 post-injury that mark the major pathohistological progression stages of neointimal formation were recorded on high-density oligonucleotide arrays. A subset of genes from microarray-based data was further studied using quantitative real time RT-PCR and in situ hybridization with sequential arterial samples from days 1 to 28 post-injury. The gene-encoded proteins were validated with Western blot. Besides temporal induction of a large cluster of genes over-represented by cell proliferation and macromolecule metabolism gene ontology categories, a fast-evolving inflammation could be demonstrated by the induction of Tgfb and other anti-inflammatory genes (e.g., C1qtnf3 (C1q and tumor necrosis factor related protein 3 (predicted))) and a shift from type 1 to 2 helper T cell response. The most significant signature of the induced neointimal profile is enrichment of genes functionally related to angiogenesis and extracellular matrix (ECM) remodeling (e.g., Spp1 (secreted phosphoprotein 1), CD44 (CD44 antigen), and Cxcl12 (chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1)). Some of the genes represent stress-responsive mesenchymal stromal cell cytokines. This study highlighted mesenchymal stromal cell cytokines-driven inflammatory extracellular matrix remodeling, as target processes for potential clinical therapeutic intervention.


Subject(s)
Angioplasty, Balloon , Carotid Arteries , Gene Expression Regulation , Angioplasty, Balloon/adverse effects , Animals , Carotid Arteries/pathology , Carotid Arteries/physiology , Carotid Arteries/surgery , Computational Biology , Gene Expression Profiling , Hyperplasia , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/metabolism , Male , Oligonucleotide Array Sequence Analysis , Oxidative Stress , Rats , Rats, Sprague-Dawley
18.
Connect Tissue Res ; 47(4): 229-34, 2006.
Article in English | MEDLINE | ID: mdl-16987755

ABSTRACT

Escherichia coli beta-galactosidase (beta-gal), encoded by the lacZ gene, has become an essential tool in studies of gene expression and function in higher eukaryotes. lac-Z is widely used as a marker gene to detect expression of transgenes or Cre recombinase driven by tissue-specific promoters. The timing and location of promoter activity is easily visualized in whole embryos or specific tissues using the cleavable, chromogenic substrate, 5-bromo-4-chloro-3-indolyl-D-galactopyranoside (X-gal). The tissue specificity of promoters in transgenic constructs is routinely tested by using a promoter of choice to drive lacZ. Alternatively, the targeted expression of Cre recombinase to perform in vivo recombination of loxP sites can be visualized by beta-gal staining in mice carrying a Cre-activated lacZ transgene, such as the ROSA26 strain. In the course of our investigations, we examined beta-gal activity in bone tissue from genetically normal mice using standard detection methodology and found very high endogenous activity in bone-resorbing osteoclasts. This was true in frozen, paraffin, and glycol methacrylate sections. X-gal staining colocalized with the osteoclast marker, tartrate-resistant acid phosphatase (TRAP). beta-gal activity was present in osteoclasts in long bones, in the mandible, and in both neonatal and more mature animals. We present this brief article as a caution to those testing genetic models of skeletal gene expression using beta-gal as a marker gene.


Subject(s)
Histocytochemistry/methods , Osteoclasts/enzymology , beta-Galactosidase/metabolism , Acid Phosphatase/metabolism , Animals , Animals, Newborn , Bone and Bones/enzymology , False Positive Reactions , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Osteoclasts/cytology , Tartrate-Resistant Acid Phosphatase
19.
J Biol Chem ; 281(33): 23598-605, 2006 Aug 18.
Article in English | MEDLINE | ID: mdl-16787916

ABSTRACT

Osteoclasts differentiate from hematopoietic mononuclear precursor cells under the control of both colony stimulating factor-1 (CSF-1, or M-CSF) and receptor activator of NF-kappaB ligand (RANKL, or TRANCE, TNFSF11) to carry out bone resorption. Using high density gene microarrays, we followed gene expression changes in long bone RNA when CSF-1 injections were used to restore osteoclast populations in the CSF-1-null toothless (csf1(tl)/csf1(tl)) osteopetrotic rat. We found that ovarian cancer G-protein-coupled receptor 1 (OGR1, or GPR68) was strongly up-regulated, rising >6-fold in vivo after 2 days of CSF-1 treatments. OGR1 is a dual membrane receptor for both protons (extracellular pH) and lysolipids. Strong induction of OGR1 mRNA was also observed by microarray, real-time RT-PCR, and immunoblotting when mouse bone marrow mononuclear cells and RAW 264.7 pre-osteoclast-like cells were treated with RANKL to induce osteoclast differentiation. Anti-OGR1 immunofluorescence showed intense labeling of RANKL-treated RAW cells. The time course of OGR1 mRNA expression suggests that OGR1 induction is early but not immediate, peaking 2 days after inducing osteoclast differentiation both in vivo and in vitro. Specific inhibition of OGR1 by anti-OGR1 antibody and by small inhibitory RNA inhibited RANKL-induced differentiation of both mouse bone marrow mononuclear cells and RAW cells in vitro, as evidenced by a decrease in tartrate-resistant acid phosphatase-positive osteoclasts. Taken together, these data indicate that OGR1 is expressed early during osteoclastogenesis both in vivo and in vitro and plays a role in osteoclast differentiation.


Subject(s)
Neoplasm Proteins/physiology , Osteoclasts/metabolism , Osteogenesis/genetics , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Animals , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Female , Macrophage Colony-Stimulating Factor/administration & dosage , Mice , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Osteoclasts/cytology , Osteopetrosis/genetics , Osteopetrosis/metabolism , Osteopetrosis/therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Mutant Strains , Receptors, G-Protein-Coupled/physiology
20.
Blood ; 107(6): 2262-70, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16304045

ABSTRACT

Osteoclasts differentiate from hematopoietic precursors under systemic and local controls. Chemokines and receptors direct leukocyte traffic throughout the body and may help regulate site-specific bone resorption. We investigated bone gene expression in vivo during rapid osteoclast differentiation induced by colony-stimulating factor 1 (CSF-1) in Csf1-null toothless (tl/tl) rats. Long-bone RNA from CSF-1-treated tl/tl rats was analyzed by high-density microarray over a time course. TRAP (tartrate-resistant acid phosphatase)-positive osteoclasts appeared on day 2, peaked on day 4, and decreased slightly on day 6, as marrow space was expanding. TRAP and cathepsin K mRNA paralleled the cell counts. We examined all chemokine and receptor mRNAs on the arrays. CCL9 was strongly induced and peaked on day 2, as did its receptor, CCR1, and regulatory receptors c-Fms (CSF-1 receptor) and RANK (receptor activator of nuclear factor kappaB). Other chemokines and receptors showed little or no significant changes. In situ hybridization and immunohistochemistry revealed CCL9 in small, immature osteoclasts on day 2 and in mature cells at later times. Anti-CCL9 antibody inhibited osteoclast differentiation in culture and significantly suppressed the osteoclast response in CSF-1-treated tl/tl rats. While various chemokines have been implicated in osteoclastogenesis in vitro, this first systematic analysis of chemokines and receptors during osteoclast differentiation in vivo highlights the key role of CCL9 in this process.


Subject(s)
Chemokines, CC/genetics , Chemokines, CC/physiology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophage Inflammatory Proteins/physiology , Osteoclasts/cytology , Receptors, Chemokine/genetics , Animals , Cathepsin K , Cathepsins/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Gene Expression Profiling , Glycoproteins/genetics , Osteopetrosis , Osteoprotegerin , RNA/analysis , Rats , Rats, Inbred Strains , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptors, CCR1 , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Tumor Necrosis Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...