Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Rev Endocrinol ; 17(6): 336-349, 2021 06.
Article in English | MEDLINE | ID: mdl-33948016

ABSTRACT

The major mineralized tissues are bone and teeth, which share several mechanisms governing their development and mineralization. This crossover includes the hormones that regulate circulating calcium and phosphate concentrations, and the genes that regulate the differentiation and transdifferentiation of cells. In developing endochondral bone and in developing teeth, parathyroid hormone-related protein (PTHrP) acts in chondrocytes to delay terminal differentiation, thereby increasing the pool of precursor cells. Chondrocytes and (in specific circumstances) pre-odontoblasts can also transdifferentiate into osteoblasts. Moreover, bone and teeth share outcomes when affected by systemic disorders of mineral homeostasis or of the extracellular matrix, and by adverse effects of treatments such as bisphosphonates and fluoride. Unlike bone, teeth have more permanent effects from systemic disorders because they are not remodelled after they are formed. This Review discusses the normal processes of bone and tooth development, followed by disorders that have effects on both bone and teeth, versus disorders that have effects in one without affecting the other. The takeaway message is that bone specialists should know when to screen for dental disorders, just as dental specialists should recognize when a tooth disorder should raise suspicions about a possible underlying bone disorder.


Subject(s)
Biomineralization/physiology , Bone Development/physiology , Bone Diseases, Developmental/metabolism , Odontogenesis/physiology , Tooth Diseases/metabolism , Animals , Bone Diseases, Developmental/pathology , Humans , Tooth Diseases/pathology
2.
Methods Mol Biol ; 816: 119-43, 2012.
Article in English | MEDLINE | ID: mdl-22130926

ABSTRACT

Osteoclasts originate from hematopoietic myeloid progenitors that differentiate into specialized multinucleated cells uniquely capable of resorbing bone in both physiological and pathological conditions. Osteoclast numbers and degradative activities increase in various inflammatory disorders of bone and certain bone oncologies, thereby causing bone loss that may weaken the skeleton, increase fracture incidence, and disturb marrow function. Many valuable insights have been obtained through the use of osteoclasts directly isolated from the bones of chickens fed a low calcium diet to enhance osteoclastogenesis and bone resorption. Particular advantages of this system include the abundance and highly resorptive nature of isolated chicken osteoclasts compared with those directly obtained from other species. After enzymatic release from the harvested bones, osteoclasts may be partially purified by density gradient sedimentation, bone substrate attachment, and/or immunomagnetic capture. Thereafter, osteoclast preparations may be analyzed, either directly or following some period of culture, to investigate their properties (biochemical, immunological, molecular, cell biological), resorptive function, and modulatory responses to various stimuli. Here, we present common procedures for the isolation, culture, and general study of chicken osteoclasts.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Osteoclasts/cytology , Animals , Antigens/analysis , Bone Resorption , Bone and Bones/cytology , Cell Movement , Cells, Cultured , Chickens , Histocytochemistry/methods , Osteoclasts/immunology , Osteoclasts/ultrastructure , Staining and Labeling/methods , Tissue Fixation/methods
3.
Methods Mol Biol ; 816: 187-202, 2012.
Article in English | MEDLINE | ID: mdl-22130930

ABSTRACT

Extensive research efforts over the years have provided us with great insights into how bone-resorbing osteoclasts (OCs) develop and function and, based on such work, valuable antiresorptive therapies have been developed to help combat the excessive bone loss that occurs in numerous skeletal disorders. The RAW 264.7 murine cell line has proven to be an important tool for in vitro studies of OC formation and function, having particular advantages over the use of OCs generated from primary bone marrow cell populations or directly isolated from murine bones. These include their ready access and availability, simple culture for this pure macrophage/pre-OC population, sensitive and rapid development into highly bone-resorptive OCs expressing hallmark OC characteristics following their RANKL stimulation, abundance of RAW cell-derived OCs that can be generated to provide large amounts of study material, relative ease of transfection for genetic and regulatory manipulation, and close correlation in characteristics, gene expression, signaling, and developmental or functional processes between RAW cell-derived OCs and OCs either directly isolated from murine bones or formed in vitro from primary bone marrow precursor cells. Here, we describe methods for the culture and RANKL-mediated differentiation of RAW cells into bone-resorptive OCs as well as procedures for their enrichment, characterization, and general use in diverse analytical assays.


Subject(s)
Cell Culture Techniques/methods , Macrophages/cytology , Osteoclasts/cytology , RANK Ligand/metabolism , Animals , Bone Resorption , Bone and Bones/cytology , Cell Differentiation , Cell Line , Macrophages/metabolism , Mice , Osteoclasts/metabolism
4.
J Bone Miner Res ; 23(12): 1983-94, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18684089

ABSTRACT

The Brtl mouse, a knock-in model for moderately severe osteogenesis imperfecta (OI), has a G349C substitution in half of type I collagen alpha1(I) chains. We studied the cellular contribution to Brtl bone properties. Brtl cortical and trabecular bone are reduced before and after puberty, with BV/TV decreased 40-45%. Brtl ObS/BS is comparable to wildtype, and Brtl and wildtype marrow generate equivalent number of colony-forming units (CFUs) at both ages. However, OcS/BS is increased in Brtl at both ages (36-45%), as are TRACP(+) cell numbers (57-47%). After puberty, Brtl ObS/BS decreases comparably to wildtype mice, but osteoblast matrix production (MAR) decreases to one half of wildtype values. In contrast, Brtl OcS falls only moderately (approximately 16%), and Brtl TRACP staining remains significantly elevated compared with wildtype. Consequently, Brtl BFR decreases from normal at 2 mo to one half of wildtype values at 6 mo. Immunohistochemistry and real-time RT-PCR show increased RANK, RANKL, and osteoprotegerin (OPG) levels in Brtl, although a normal RANKL/OPG ratio is maintained. TRACP(+) precursors are markedly elevated in Brtl marrow cultures and form more osteoclasts, suggesting that osteoclast increases arise from more RANK-expressing precursors. We conclude that osteoblasts and osteoclasts are unsynchronized in Brtl bone. This cellular imbalance results in declining BFR as Brtl ages, consistent with reduced femoral geometry. The disparity in cellular number and function results from poorly functioning osteoblasts in addition to increased RANK-expressing precursors that respond to normal RANKL/OPG ratios to generate more bone-resorbing osteoclasts. Interruption of the stimulus that increases osteoclast precursors may lead to novel OI therapies.


Subject(s)
Bone and Bones/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis Imperfecta/genetics , Amino Acids/chemistry , Animals , Bone Marrow Cells/cytology , Disease Models, Animal , Fibroblasts/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RANK Ligand/metabolism , Stem Cells
5.
J Biol Chem ; 281(23): 15809-20, 2006 Jun 09.
Article in English | MEDLINE | ID: mdl-16613848

ABSTRACT

Nitric oxide (NO) is a multifunctional signaling molecule and a key vasculoprotective and potential osteoprotective factor. NO regulates normal bone remodeling and pathological bone loss in part through affecting the recruitment, formation, and activity of bone-resorbing osteoclasts. Using murine RAW 264.7 and primary bone marrow cells or osteoclasts formed from them by receptor activator of NF-kappaB ligand (RANKL) differentiation, we found that inducible nitric-oxide synthase (iNOS) expression and NO generation were stimulated by interferon (IFN)-gamma or lipopolysaccharide, but not by interleukin-1 or tumor necrosis factor-alpha. Surprisingly, iNOS expression and NO release were also triggered by RANKL. This response was time- and dose-dependent, required NF-kappaB activation and new protein synthesis, and was specifically blocked by the RANKL decoy receptor osteoprotegerin. Preventing RANKL-induced NO (via iNOS-selective inhibition or use of marrow cells from iNOS-/- mice) increased osteoclast formation and bone pit resorption, indicating that such NO normally restrains RANKL-mediated osteoclastogenesis. Additional studies suggested that RANKL-induced NO inhibition of osteoclast formation does not occur via NO activation of a cGMP pathway. Because IFN-beta is also a RANKL-induced autocrine negative feedback inhibitor that limits osteoclastogenesis, we investigated whether IFN-beta is involved in this novel RANKL/iNOS/NO autoregulatory pathway. IFN-beta was induced by RANKL and stimulated iNOS expression and NO release, and a neutralizing antibody to IFN-beta inhibited iNOS/NO elevation in response to RANKL, thereby enhancing osteoclast formation. Thus, RANKL-induced IFN-beta triggers iNOS/NO as an important negative feedback signal during osteoclastogenesis. Specifically targeting this novel autoregulatory pathway may provide new therapeutic approaches to combat various osteolytic bone diseases.


Subject(s)
Bone Resorption , Carrier Proteins/physiology , Interferon-beta/physiology , Membrane Glycoproteins/physiology , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/biosynthesis , Osteoclasts/metabolism , Animals , Base Sequence , Cell Line , DNA Primers , Enzyme Inhibitors/pharmacology , Interferon-beta/biosynthesis , Mice , Nitric Oxide Synthase Type II/antagonists & inhibitors , Osteoclasts/enzymology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Reverse Transcriptase Polymerase Chain Reaction
6.
J Bone Miner Res ; 21(2): 193-206, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16418775

ABSTRACT

UNLABELLED: Circulating pre-OCs may be recruited to locally inflamed sites through specific interactions with activated microvasculature. We found that HMVECs stimulated the adhesion and TEM of circulating pre-OCs, in an ICAM-1- and CD44-dependent manner, leading to greater RANKL-induced OC formation and bone pit resorption. INTRODUCTION: Inflammation is critical for healing processes but causes severe tissue destruction when chronic. Local osteoclast (OC) formation and bone resorption may increase at inflammatory sites through multiple mechanisms, including direct stimulation by inflamed microvasculature of circulating OC precursor (pre-OC) migration through a blood vessel barrier into bone or joint tissue. How this might occur is not yet well understood. MATERIALS AND METHODS: Cytokine-activated human microvascular endothelial cell (HMVEC) monolayers, with or without IL-1 and TNF-alpha preactivation (24 h), were incubated in adhesion (1-3 h) or porous transwell transendothelial migration (TEM; 3 h) assays with human peripheral blood mononuclear cells (hPBMCs) or CD14+ monocyte or CD14- lymphocyte subsets. The number of cells that adhered or transmigrated, and their ability to thereafter develop with macrophage-colony stimulating factor (M-CSF) + RANKL into bone pit-resorbing OCs, were analyzed. Immunostaining and neutralizing antibodies to key cell adhesion molecules were used to determine their potential involvement in stimulated CD14+ monocyte TEM. RESULTS: M-CSF + RANKL caused OC and bone pit formation only from hPBMCs and CD14+ cells but not CD14- cells. Adhesion of hPBMCs or CD14+ cells but not CD14- cells was stimulated by cytokine preactivation of HMVECs and led to the full capture of all circulating pre-OCs capable of developing into OCs. Cytokine-preactivated HMVECs also promoted the postadhesion TEM of hPBMCs and CD14+ populations, resulting in markedly greater OC formation and bone pit resorption by transmigrated cells. Immunodetectable vascular cell adhesion molecule (VCAM-1), intercellular adhesion molecule (ICAM-1), and CD44 levels increased on cytokine-treated HMVEC surfaces, and neutralizing antibodies to ICAM-1 or CD44, but not VCAM-1 or platelet endothelial cell adhesion molecule (PECAM-1), inhibited stimulated CD14+ cell TEM through activated HMVECs. CONCLUSIONS: This is the first demonstration that cytokine-activated HMVECs efficiently capture and promote the TEM of circulating pre-OCs capable of differentiating into bone-resorbing OCs. Thus, direct pre-OC recruitment by activated microvasculature at inflammatory sites may significantly contribute to normal OC bone remodeling during fracture healing or exacerbate pathological bone loss in various chronic inflammatory disorders.


Subject(s)
Bone Remodeling , Cell Movement , Endothelium, Vascular/drug effects , Interleukin-1/pharmacology , Osteoclasts/cytology , Tumor Necrosis Factor-alpha/pharmacology , Bone Resorption , Capillaries/cytology , Capillaries/drug effects , Carrier Proteins/pharmacology , Cell Adhesion , Cell Differentiation , Female , Humans , Hyaluronan Receptors/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lipopolysaccharide Receptors/analysis , Macrophage Colony-Stimulating Factor/pharmacology , Membrane Glycoproteins/pharmacology , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B
7.
J Cell Physiol ; 204(3): 871-80, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15828028

ABSTRACT

The osteoclast is a highly polarized multinucleated cell that resorbs bone. Using high resolution immunofluorescence microscopy, we demonstrated that all nuclei of an osteoclast are transcriptionally active. Each nucleus within the osteoclast contains punctately organized microenvironments where regulatory complexes that support transcriptional and post-transcriptional control reside. Functional equivalency of osteoclast nuclei is reflected by similar representation of regulatory proteins that support ribosomal RNA synthesis (nucleolin), mRNA transcription (RNA polymerase II, bromouridine triphosphate), processing of gene transcripts (SC35), signal transduction (NF-kappaB), and phenotypic gene expression (Runx1). Our results establish that gene regulatory machinery is architecturally associated and compartmentalized within intranuclear microenvironments of the multiple nuclei of osteoclasts to support physiologically responsive modifications in cellular structural and functional properties.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Carrier Proteins/pharmacology , Cell Differentiation/drug effects , Cell Lineage , Cell Nucleus/drug effects , Cells, Cultured , Core Binding Factor Alpha 2 Subunit , Down-Regulation , Male , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Nuclear Matrix/metabolism , Organ Specificity , Osteoclasts/drug effects , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B
8.
Bone ; 36(5): 840-53, 2005 May.
Article in English | MEDLINE | ID: mdl-15794931

ABSTRACT

Osteoclasts (Oc) derive from hematopoietic precursors present in the circulation and bone marrow, and they differentiate into multinucleated bone-resorbing cells in response to the dual essential signals receptor activator of NF-kappaB ligand (RANKL) and macrophage-colony stimulating factor (M-CSF) primarily provided by bone marrow stromal cells (BMSC) and osteoblasts (Ob). However, little is known about signals that direct Oc precursors from the circulation into bone or control their migration within the marrow. Stromal cell-derived factor-1 (SDF-1 or CXCL12) is a chemokine highly expressed by bone endothelium, BMSC, and immature Ob that is essential for the normal homing, early development, and survival of various hematopoietic progenitor cells. We investigated whether SDF-1 and its unique chemokine receptor CXCR4 were involved in regulating human Oc precursor chemotaxis, development, function, or survival. CXCR4 was highly expressed by freshly isolated human monocyte (MN) populations, in vitro generated Oc and Oc-like cells, and mature Oc isolated from human femoral bones. SDF-1 markedly stimulated the chemotactic recruitment of circulating human MN capable of generating bone-resorptive Oc, leading to a 4-fold increase in Oc formation and greater bone pit resorption after their M-CSF + RANKL induced differentiation compared to spontaneously migrating cells. SDF-1 also directly promoted early (but not later) stages of Oc development via stimulating precursor cell numbers, multinucleated cell fusion, increased cell size, and tartrate-resistant acid phosphatase (TRAP) activity in a similar, but non-additive, fashion to M-CSF + RANKL. While SDF-1 did not cause full development of bone-resorbing Oc or stimulate the resorptive function of mature Oc directly, it also did not interfere with any actions promoted by M-CSF + RANKL. In mature human Oc, SDF-1 proved equally as effective as M-CSF + RANKL for preventing Oc apoptosis induced by cytokine withdrawal. In both cases, Oc survival was accompanied by analogous rises in the mRNA ratios for anti-apoptotic Bcl-xL and Bfl-1 relative to pro-apoptotic Bax, and by marked protein suppression of the critical pro-apoptotic signal Bim. These findings demonstrate for the first time that SDF-1 chemoattracts circulating human Oc precursors capable of developing into bone-resorptive Oc, and that it can stimulate MN cell fusion and TRAP activity, mimic M-CSF + RANKL in early osteoclastogenic effects, substitute for M-CSF + RANKL in maintaining the survival of mature human Oc, and suppress Oc expression of Bim protein. Thus, high levels of SDF-1 produced by bone endothelium, BMSC, and Ob may selectively target circulating Oc precursors into bone and stimulate their marrow migration into suitable perivascular stromal sites for their early development, RANKL differentiation, and survival. Consequently, SDF-1 may be a key factor linking bone vascular cells, BMSC, Ob, and Oc in the normal homeostatic regulation of bone development and remodeling.


Subject(s)
Cell Survival/physiology , Chemokines, CXC/metabolism , Chemotaxis/physiology , Osteoclasts/cytology , Receptors, CXCR4/metabolism , Blotting, Western , Cells, Cultured , Chemokine CXCL12 , Chemokines, CXC/physiology , Humans , Protein Binding , Recombinant Proteins/metabolism
9.
J Bone Miner Res ; 19(12): 2065-77, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15537451

ABSTRACT

UNLABELLED: Chemoattractants that recruit OC precursors to locally inflamed sites of resorption are not well known. A chemokine receptor, CCR1, was expressed in OC precursors and elevated in mature OCs, and its ligands promoted OC precursor recruitment, RANKL development, and OC motility. Cytokines induced OB release of such chemokines, which may therefore significantly contribute to inflammatory bone loss. INTRODUCTION: Chemokines, primarily of two major (CXC, CC) families, are essential signals for the trafficking and localization of circulating hematopoietic cells into tissues. However, little is known about their potential roles in osteoclast (OC) recruitment, development, or function. Previously, we analyzed CXC receptors in murine OC precursors and found high expression of CXCR4 that mediated their stromal-derived factor-1(SDF-1)-induced chemotaxis and collagen invasion. Here, we investigated if CC receptors and ligands, which are elevated in inflammatory and other osteolytic diseases, also play important roles in the recruitment, formation, or activity of murine bone-resorptive OCs. MATERIALS AND METHODS: CC chemokine receptor (CCR) mRNA expression was analyzed during OC formation induced by RANKL in murine RAW 264.7 cells and primary marrow cells. Corresponding CC chemokines were tested for their ability to elicit precursor chemotaxis or OC development, or to influence motility, bone resorption, adhesion, or survival in RANKL-differentiated OCs. Constitutive and inflammatory cytokine-induced release of the chemokines macrophage inflammatory protein-1alpha (MIP-1alpha) and regulated on activation, normal T-cell expressed and secreted (RANTES) was measured by ELISA for OCs, osteoblasts (OBs), and their precursor cells. RESULTS: CCR1 was expressed in murine marrow cells, the most prominent CCR in RAW cells, and upregulated by RANKL in marrow or RAW cells. Chemokines that bind CCR1 (MIP-1alpha, RANTES, and monocyte chemoattractant protein-3 [MCP-3]) were produced to varying degrees by murine OCs, OBs, and their precursors, and markedly increased by interleukin (IL)-1alpha and TNFalpha in differentiating OBs. RANTES, and especially MIP-1alpha, increased mature OC motility, but did not alter OC resorption activity, adhesion, or survival. All three chemokines stimulated chemotaxis of marrow or RAW cell precursors, leading to the greater formation of OCs (in number and size) after RANKL development of such chemoattracted marrow cells. All three chemokines also directly and dramatically enhanced OC formation in marrow cultures, through a pathway dependent on the presence of RANKL but without altering RANK expression. CONCLUSIONS: Pathological increases in secretion of these chemokines from activated OBs or other cells may potently stimulate the chemotactic recruitment and RANKL formation of bone-resorptive OCs, thereby exacerbating local osteolysis in multiple skeletal diseases.


Subject(s)
Bone Resorption , Carrier Proteins/metabolism , Cytokines/metabolism , Membrane Glycoproteins/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Receptors, Chemokine/physiology , Animals , Bone Diseases/metabolism , Bone and Bones/metabolism , Cell Movement , Cells, Cultured , Centrifugation, Density Gradient , Chemokine CCL3 , Chemokine CCL4 , Chemokine CCL5/metabolism , Chemokine CCL7 , Chemokine CXCL12 , Chemokines/metabolism , Chemokines, CXC/metabolism , Chemotaxis , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Inflammation , Ligands , Macrophage Inflammatory Proteins/metabolism , Mice , Models, Biological , Monocyte Chemoattractant Proteins/metabolism , RANK Ligand , RNA/chemistry , RNA, Messenger/metabolism , Receptor Activator of Nuclear Factor-kappa B , Receptors, CCR1 , Receptors, Chemokine/metabolism , Ribonucleases/chemistry , Up-Regulation
10.
Connect Tissue Res ; 44 Suppl 1: 79-84, 2003.
Article in English | MEDLINE | ID: mdl-12952178

ABSTRACT

Although chemokines play essential roles in the trafficking and homing of many circulating hematopoietic cell types, their potential influences on osteoclast (OC) recruitment or bone remodeling are not well known. Therefore, chemokine receptor expression was analyzed by RNase protection assay during OC formation induced by RANKL in a murine mononuclear cell line (RAW 264.7). Relatively high CXCR4 expression was detected in RAW cells (pre-OCs), whereas CXCR4 levels were downregulated during RAW-OC development. SDF-1, the unique ligand for CXCR4, stimulated RAW cell production of matrix metalloproteinase (MMP)-9 activity, a matrix-degrading enzyme essential for pre-OC migration into the developing bone marrow cavity. Induced MMP-9 activity in RAW cells was associated with their increased MMP-dependent transmigration through a collagen gel in response to SDF-1. We conclude that SDF-1 stimulation of MMP-9 activity in pre-OCs may be a key aspect of their recruitment to bone and migration within the marrow to sites for OC differentiation and bone resorption.


Subject(s)
Cell Movement/drug effects , Chemokines, CXC/pharmacology , Matrix Metalloproteinase 9/biosynthesis , Osteoclasts/drug effects , Stem Cells/drug effects , Animals , Carrier Proteins/pharmacology , Cell Differentiation/drug effects , Cell Line , Chemokine CXCL12 , Macrophages , Membrane Glycoproteins/pharmacology , Mice , Osteoclasts/cytology , Osteoclasts/enzymology , RANK Ligand , RNA, Messenger/metabolism , Receptor Activator of Nuclear Factor-kappa B , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/genetics , Stem Cells/cytology , Stem Cells/enzymology , Up-Regulation
11.
J Bone Miner Res ; 18(8): 1404-18, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12929930

ABSTRACT

UNLABELLED: Signals targeting OCs to bone and resorption sites are not well characterized. A chemoattractant receptor (CXCR4), highly expressed in murine OC precursors, mediated their chemokine (SDF-1)-induced chemoattraction, collagen transmigration, and MMP-9 expression. Thus, bone vascular and stromal SDF-1 may direct OC precursors into bone and marrow sites for development and bone resorption. INTRODUCTION: Although chemokines are essential for trafficking and homing of circulating hematopoietic cells under normal and pathological conditions, their potential roles in osteoclast (OC) recruitment or function are generally unknown. CXCR4 and its unique ligand, stromal cell-derived factor-1 (SDF-1), critically control the matrix metalloproteinase (MMP)-dependent targeting of hematopoietic cells into bone and within the marrow microenvironment. Therefore, SDF-1/CXCR4 may regulate OC precursor recruitment to sites for development and activation. METHODS: Chemokine receptor mRNA expression was analyzed during OC formation induced by RANKL in murine RAW 264.7 cells. SDF-1 versus RANKL effects on chemotaxis, transcollagen migration, MMP-9 expression and activity, OC development, and bone resorption were evaluated in RAW cells or RAW-OCs. RESULTS: CXCR4 was highly expressed in RAW cells and downregulated during their RANKL development into bone-resorptive RAW-OCs. SDF-1, but not RANKL, elicited RAW cell chemotaxis. Conversely, RANKL, but not SDF-1, promoted RAW-OC development, TRAP activity, cathepsin K expression, and bone pit resorption, and SDF-1 did not modify these RANKL responses. Both SDF-1 and RANKL increased MMP-9, a matrix-degrading enzyme essential for OC precursor migration into developing bone marrow cavities, and increased transcollagen migration of RAW cells in a MMP-dependent manner. SDF-1 also upregulated MMP-9 in various primary murine OC precursor cells. Because RANKL induced a higher, more sustained expression of MMP-9 in RAW cells than did SDF-1, MMP-9 may have an additional role in mature OCs. Consistent with this, MMP-9 upregulation during RANKL-induced RAW-OC development was necessary for initiation of bone pit resorption. CONCLUSIONS: SDF-1, a chemokine highly expressed by bone vascular endothelial and marrow stromal cells, may be a key signal for the selective attraction of circulating OC precursors into bone and their migration within marrow to appropriate perivascular stromal sites for RANKL differentiation into resorptive OCs. Thus, SDF-1 and RANKL likely serve complementary physiological functions, partly mediated through increases in MMP-9, to coordinate stages of OC precursor recruitment, development, and function.


Subject(s)
Chemokines, CXC/pharmacology , Chemotaxis/drug effects , Collagen/metabolism , Matrix Metalloproteinase 9/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Stem Cells/drug effects , Animals , Bone Resorption , Carrier Proteins/pharmacology , Chemokine CXCL12 , Enzyme Induction/drug effects , Matrix Metalloproteinase 2/metabolism , Membrane Glycoproteins/pharmacology , Mice , Osteoclasts/cytology , Osteoclasts/enzymology , RANK Ligand , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Activator of Nuclear Factor-kappa B , Receptors, Chemokine/genetics , Stem Cells/cytology , Stem Cells/enzymology , Stem Cells/metabolism
14.
J Bone Miner Res ; 17(10): 1859-71, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12369790

ABSTRACT

Increased local osteoclast (OC)-mediated bone resorption coincides with angiogenesis in normal bone development and fracture repair, as well as in pathological disorders such as tumor-associated osteolysis and inflammatory-related rheumatoid arthritis or periodontal disease. Angiogenic stimulation causes recruitment, activation, adhesion, transmigration, and differentiation of hematopoietic cells which may therefore enable greater numbers of pre-OC to emigrate from the circulation and develop into bone-resorptive OCs. A chick chorioallantoic membrane (CAM) model, involving coimplantation of a stimulus in an agarose plug directly adjacent to a bone chip was used to investigate if a potent angiogenic stimulator, basic fibroblast growth factor (bFGF), could promote OC recruitment, differentiation, and resorption in vivo. Angiogenesis elicited by bFGF on the CAM was accompanied by increased OC formation and bone pit resorption (both overall and on a per OC basis) on the bone implants in vivo. In complementary in vitro assays, bFGF did not directly stimulate avian OC development from bone marrow mononuclear cell precursors, consistent with their low mRNA expression of the four avian signaling FGF receptors (FGFR)-1, FGFR-2, FGFR-3, and FGFR-like embryonic kinase (FREK). In contrast, bFGF activated isolated avian OC bone pit resorption via mechanisms inhibited by a selective cyclo-oxygenase (COX)-2 prostaglandin inhibitor (NS-398) or p42/p44 MAPK activation inhibitor (PD98059), consistent with a relatively high expression of FGFR-1 by differentiated avian OCs. Thus, bFGF may sensitively regulate local bone resorption and remodeling through direct and indirect mechanisms that promote angiogenesis and OC recruitment, formation, differentiation, and activated bone pit resorption. The potential for bFGF to coinduce angiogenesis and OC bone remodeling may find clinical applications in reconstructive surgery, fracture repair, or the treatment of avascular necrosis. Alternatively, inhibiting such bFGF-dependent processes may aid in the treatment of inflammatory-related or metastatic bone loss.


Subject(s)
Allantois/drug effects , Bone Resorption/chemically induced , Chorion/drug effects , Fibroblast Growth Factor 2/pharmacology , Neovascularization, Physiologic/drug effects , Osteoclasts/drug effects , Animals , Cell Differentiation/drug effects , Cells, Cultured/drug effects , Chick Embryo , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Drug Implants , Flavonoids/pharmacology , Gene Expression Regulation, Developmental/drug effects , Humans , Isoenzymes/antagonists & inhibitors , Membrane Proteins , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nitrobenzenes/pharmacology , Prostaglandin-Endoperoxide Synthases , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 4 , Receptors, Fibroblast Growth Factor/biosynthesis , Receptors, Fibroblast Growth Factor/genetics , Recombinant Proteins/pharmacology , Sulfonamides/pharmacology
15.
Cytometry ; 50(5): 261-6, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12360576

ABSTRACT

BACKGROUND: Osteoclast cell function relates to bone resorption. Isolation and characterization of these cells from in vivo sources remain difficult. The aim of this study was to show the feasibility of using flow cytometry to identify and characterize human mature osteoclasts obtained from bone tissues. METHODS: Bone femoral heads obtained as discarded surgical material were used. To check the nature of 121F(+) (a monoclonal antibody specific for human osteoclasts) cells by flow cytometry, we used laser scanning cytometry to analyze simultaneously the immunophenotype and DNA cell content of osteoclast-like cell-enriched bone samples. RESULTS: Results were compared with conventional morphologic and cytochemical studies. The percentage of cells that showed both cytochemical (tartrate-resistant acid phosphatase [TRAP](+)) and immunophenotypic (121F(+)) osteoclast-associated characteristics was very similar (12.5 +/- 6.2 versus 14.7 +/- 11.7; P = 0.46). Laser scanning cytometry showed that 121F(+) cells were bigger (P = 0.04) and they had a higher DNA cell content (P = 0.04) and more nuclei per cell (P = 0.04) than the 121F(-) cells present in the same sample. DISCUSSION: This study relied on the combined use of the 121F(+) antibody and different cytometry-based techniques to characterize the osteoclast populations from human bone.


Subject(s)
Bone and Bones/cytology , Bone and Bones/immunology , Cell Separation/methods , Flow Cytometry/methods , Immunophenotyping/methods , Osteoclasts/cytology , Osteoclasts/immunology , Acid Phosphatase , Antibodies, Monoclonal , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cell Separation/instrumentation , Chromatin/metabolism , Chromatin/ultrastructure , DNA/metabolism , Flow Cytometry/instrumentation , Histocytochemistry , Humans , Immunophenotyping/instrumentation , Isoenzymes , Microscopy, Confocal/methods , Reproducibility of Results , Tartrate-Resistant Acid Phosphatase
16.
J Biol Chem ; 277(21): 18840-8, 2002 May 24.
Article in English | MEDLINE | ID: mdl-11886849

ABSTRACT

Osteoclasts or their precursors interact with the glycoprotein-enriched matrix of bone during extravasation from the vasculature, and upon attachment prior to resorption. Reverse transcriptase-PCR studies showed that two new alternatively spliced forms of chicken galectin-3, termed Gal-3TM1 and Gal-3TR1, were enriched and preferentially expressed in highly purified chicken osteoclast-like cells. Gal-3TM1 and Gal-3TR1 mRNA were also detected in chicken intestinal tissue, but not in kidney, liver, or lung. Gal-3TM1 and Gal-3TR1 messages both contain an open reading frame encoding a predicted 70-amino acid TM1 sequence inserted between the N-terminal Gly/Pro repeat domain and the carbohydrate recognition domain (exons 3 and 4). Gal-3TR1 mRNA contains an additional 241-bp sequence, which encodes a truncated open reading frame between the 4th and 5th exons, and, whose translation is expected to terminate within the carbohydrate recognition domain encompassing exons 4, 5, and 6. Immunoblotting and affinity chromatography showed that purified osteoclast preparations and intestinal homogenates contained a 36-kDa lactose-binding galectin. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometric analyses on chymotryptic peptides from the 36-kDa lectin confirmed its identity as Gal-3TM1. The TM1 insert contains a single transmembrane-spanning region and a leucine zipper-like stalk domain that is predicted to position the intact carbohydrate recognition domain of Gal-3TM1 on the exterior surface of the plasma membrane. Immunofluorescent staining of chicken osteoclasts confirmed the expression of Gal-3TM1 at the plasma membrane. Gal-3TM1 is the first example of a galectin superfamily member capable of being expressed as a soluble protein and as a transmembrane protein.


Subject(s)
Alternative Splicing , Antigens, Differentiation/metabolism , Galactosides/metabolism , Leucine Zippers , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Antigens, Differentiation/chemistry , Antigens, Differentiation/genetics , Base Sequence , Cells, Cultured , Cloning, Molecular , DNA, Complementary , Galectin 3 , Humans , Intestinal Mucosa/metabolism , Molecular Sequence Data , Osteoclasts/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...