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










Publication year range
1.
Front Biosci (Landmark Ed) ; 16(8): 2812-902, 2011 06 01.
Article in English | MEDLINE | ID: mdl-21622210

ABSTRACT

Matrix vesicles (MVs) induce calcification during endochondral bone formation. Experimental methods for structural, compositional, and functional analysis of MVs are reviewed. MV proteins, enzymes, receptors, transporters, regulators, lipids and electrolytes are detailed. MV formation is considered from both structural and biochemical perspectives. Confocal imaging of Ca(2+) and H(+) were used to depict how living chondrocytes form MVs. Biochemical studies revealed that coordinated mitochondrial Ca(2+) and Pi metabolism produce MVs containing a nucleational complex (NC) of amorphous calcium phosphate, phosphatidylserine and annexin A5--all critical to the mechanism of mineral nucleation. Reconstitution of the NC and modeling with unilamellar vesicles reveal how the NC transforms into octacalcium phosphate, regulated by Mg(2+), Zn(2+) and annexin A5. Extravasation of intravesicular mineral is mediated by phospholipases and tissue-nonspecific alkaline phosphatase (TNAP). In the extravesicular matrix, hydroxyapatite crystal propagation is enhanced by cartilage collagens and TNAP, which destroys inhibitory PPi, and by metalloproteases that degrade proteoglycans. Other proteins also modulate mineral formation. Recent findings from single and multiple gene knockouts of TNAP, NPP1, ANK, PHOSPHO1, and Annexin A5 are reviewed.


Subject(s)
Calcification, Physiologic , Extracellular Matrix/physiology , Extracellular Matrix/ultrastructure , Animals , Bone Development/physiology , Calcium/metabolism , Cell Fractionation , Chondrocytes/physiology , Chondrocytes/ultrastructure , Cytological Techniques , Cytoskeletal Proteins/physiology , Extracellular Matrix Proteins/physiology , Humans , Lipid Metabolism , Osteogenesis/physiology , Proteomics
2.
Microsc Microanal ; 15(6): 545-57, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19758474

ABSTRACT

The differential accumulation of fluorescent molecules in tumorigenic versus normal cells is a well-reported phenomenon and is the basis for photodiagnostic therapy. Through the use of confocal microscopy, the kinetic uptake and accumulation of fusarochromanone (FC101) was determined in two lines of living tumorigenic cells of mesenchymal-epithelial origin and normal fibroblast cells. Like other fluorescent cationic molecules, FC101 showed increased accumulation in tumorigenic cells; however, unlike other molecules, it appeared to be accumulated in a time-dependent manner. Also, unlike traditional fluorescent cationic molecules, FC101, a potent inhibitor of cell growth, showed preferential inhibition of tumorigenic B-16 melanoma cells and MCF7 cells derived from breast cancer adenocarcinoma when compared to normal cardiac fibroblasts. Further analysis of FC101's physicochemical properties using both experimentally obtained and simulated values revealed the likelihood of membrane permeation and oral bioavailability of the compound. These physicochemical properties of FC101 were also used to predict its intracellular localization lending credence to data observed by confocal microscopy.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cell Membrane Permeability , Chromones/pharmacokinetics , Fibroblasts/metabolism , Fluorescent Dyes/pharmacokinetics , Neoplasms/metabolism , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Chromones/chemistry , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibroblasts/drug effects , Fluorescent Dyes/chemistry , Humans , Kinetics , Mice , Neoplasms/drug therapy
3.
J Inorg Biochem ; 103(7): 948-62, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19477528

ABSTRACT

Mg(2+) and Zn(2+) are present in the mineral of matrix vesicles (MVs) and biological apatites, and are known to influence the onset and progression of mineral formation by amorphous calcium phosphate (ACP) and hydroxyapatite (HAP). However, neither has been studied systematically for its effect on mineral formation by phosphatidylserine-Ca(2+)-Pi complexes (PS-CPLX), an important constituent of the MV nucleation core. Presented here are studies on the effects of increasing levels of Mg(2+) and Zn(2+) on the process of mineral formation, either when present in synthetic cartilage lymph (SCL), or when incorporated during the formation of PS-CPLX. Pure HAP and PS-CPLX proved to be powerful nucleators, but ACP took much longer to induce mineral formation. In SCL, Mg(2+) and Zn(2+) had significantly different inhibitory effects on the onset and amount of mineral formation; HAP and PS-CPLX were less affected than ACP. Mg(2+) and Zn(2+) caused similar reductions in the rate and length of rapid mineral formation, but Zn(2+) was a more potent inhibitor on a molar basis. When incorporated into PS-CPLX, Mg(2+) and Zn(2+) caused significantly different effects than when present in SCL. Even low, subphysiological levels of Mg(2+) altered the inherent structure of PS-CPLX and markedly reduced its ability to induce and propagate mineral formation. Incorporated Zn(2+) caused significantly less effect, low (<20 microM) levels causing almost no inhibition. Levels of Zn(2+) present in MVs do not appear to inhibit their nucleational activity.


Subject(s)
Calcification, Physiologic/drug effects , Calcium Phosphates/metabolism , Durapatite/metabolism , Magnesium/pharmacology , Phosphatidylserines/metabolism , Zinc/pharmacology , Calcification, Physiologic/physiology , Calcium Phosphates/chemical synthesis , Durapatite/chemical synthesis
4.
J Biol Chem ; 283(15): 9737-48, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18250169

ABSTRACT

Matrix vesicles (MVs) in the growth plate bind to cartilage collagens and initiate mineralization of the extracellular matrix. Native MVs have been shown to contain a nucleational core responsible for mineral formation that is comprised of Mg(2+)-containing amorphous calcium phosphate and lipid-calcium-phosphate complexes (CPLXs) and the lipid-dependent Ca(2+)-binding proteins, especially annexin-5 (Anx-5), which greatly enhances mineral formation. Incorporation of non-Ca(2+)-binding MV lipids impedes mineral formation by phosphatidylserine (PS)-CPLX. In this study, nucleators based on amorphous calcium phosphate (with or without Anx-5) were prepared with PS alone, PS + phosphatidylethanolamine (PE), or PS + PE and other MV lipids. These were incubated in synthetic cartilage lymph containing no collagen or containing type II or type X collagen. Dilution of PS with PE and other MV lipids progressively retarded nucleation. Incorporation of Anx-5 restored nucleational activity to the PS:PE CPLX; thus PS and Anx-5 proved to be critical for nucleation of mineral. Without Anx-5, induction of mineral formation was slow unless high levels of Ca(2+) were used. The presence of type II collagen in synthetic cartilage lymph improved both the rate and amount of mineral formation but did not enhance nucleation. This stimulatory effect required the presence of the nonhelical telopeptides. Although type X collagen slowed induction, it also increased the rate and amount of mineral formation. Both type II and X collagens markedly increased mineral formation by the MV-like CPLX, requiring Anx-5 to do so. Thus, Anx-5 enhances nucleation by the CPLXs and couples this to propagation of mineral formation by the cartilage collagens.


Subject(s)
Annexin A5/chemistry , Calcification, Physiologic , Calcium Phosphates/chemistry , Collagen Type II/chemistry , Collagen Type X/chemistry , Lipids/chemistry , Animals , Cartilage/chemistry , Humans
5.
J Biol Chem ; 283(7): 3827-38, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18077457

ABSTRACT

The nucleational core of matrix vesicles contains a complex (CPLX) of phosphatidylserine (PS), Ca(2+), and inorganic phosphate (P(i)) that is important to both normal and pathological calcification. Factors required for PS-CPLX formation and nucleational activity were studied using in vitro model systems and molecular dynamic simulations. Ca(2+) levels required for and rates of PS-CPLX formation were monitored by light scattering at 340 nm, assessing changes in amount and particle size. Fourier transform infrared spectroscopy was used to explore changes in chemical structure and composition. Washing with pH 5 buffer was used to examine the role of amorphous calcium phosphate in CPLX nucleational activity, which was assessed by incubation in synthetic cartilage lymph with varied pH values. Addition of 4 Ca(2+)/PS was minimally required to form viable complexes. During the critical first 10-min reaction period, rapid reduction in particle size signaled changes in PS-CPLX structure. Fourier transform infrared spectroscopy revealed increasing mineral phosphate that became progressively deprotonated to PO(4)(3-). This Ca(2+)-mediated effect was mimicked in part by increasing the Ca(2+)/PS reaction ratio. Molecular dynamic simulations provided key insight into initial interactions between Ca(2+) and P(i) and the carboxyl, amino, and phosphodiester groups of PS. Deduced interatomic distances agreed closely with previous radial distribution function x-ray-absorption fine structure measurements, except for an elongated Ca(2+)-N distance, suggesting additional changes in atomic structure during the critical 10-min ripening period. These findings clarify the process of PS-CPLX formation, reveal details of its structure, and provide insight into its role as a nucleator of crystalline calcium phosphate mineral formation.


Subject(s)
Calcium/chemistry , Phosphates/chemistry , Phosphatidylserines/chemistry , Models, Molecular , Molecular Structure , Spectroscopy, Fourier Transform Infrared
6.
Anticancer Drugs ; 18(8): 897-904, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17667595

ABSTRACT

Fusarochromanone is a toxic metabolite produced by Fusarium equiseti, a fungus present in decaying cereal plants in northern latitudes; it has been detected in various food grains. Fusarochromanone has been shown to have both stimulatory and inhibitory effects on various mammalian cells, depending on the concentration used. Whether these cytotoxic effects can be used in the clinical treatment of tumors remains to be established. Here, we evaluated the effects of fusarochromanone on the growth of human melanoma cells both in vitro and in vivo. In vitro, low concentrations (0.1-1 nmol/l) of fusarochromanone were found to be cytotoxic to many melanoma cell lines. In contrast, growth of normal melanocytes was inhibited only at much higher fusarochromanone concentrations (100-200 nmol/l). In vivo, the growth of melanoma cells implanted subcutaneously in immuno-compromised mice was significantly (P<0.05) reduced by daily administration of fusarochromanone. Immunohistological analyses indicated a significant (P<0.05) increase in the expression of active caspase-3 in tumor masses of mice treated with fusarochromanone, compared with controls. Together, these observations show that fusarochromanone increased apoptosis of tumor cells and reduced tumor growth in vivo. Therefore, the effects of fusarochromanone warrant further investigation as an adjuvant molecule to prevent growth and recurrence of melanomas.


Subject(s)
Antineoplastic Agents , Chromones/pharmacology , Melanoma/drug therapy , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Chromones/toxicity , Humans , Immunohistochemistry , Indicators and Reagents , Melanoma/blood supply , Melanoma/pathology , Mice , Mice, SCID , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Protein Synthesis Inhibitors/pharmacology , Thymidine/metabolism
7.
J Biol Chem ; 282(36): 26035-45, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17613532

ABSTRACT

Annexins A5, A2, and A6 (Anx-A5, -A2, and -A6) are quantitatively major proteins of the matrix vesicle nucleational core that is responsible for mineral formation. Anx-A5 significantly activated the induction and propagation of mineral formation when incorporated into synthetic nucleation complexes made of amorphous calcium phosphate (ACP) and Anx-A5 or of phosphatidylserine (PS) plus ACP (PS-CPLX) and Anx-A5. Incorporation of Anx-A5 markedly shortened the induction time, greatly increasing the rate and overall amount of mineral formed when incubated in synthetic cartilage lymph. Constructed by the addition of Ca(2+) to PS, emulsions prepared in an intracellular phosphate buffer matched in ionic composition to the intracellular fluid of growth plate chondrocytes, these biomimetic PS-CPLX nucleators had little nucleational activity. However, incorporation of Anx-A5 transformed them into potent nucleators, with significantly greater activity than those made from ACP without PS. The ability of Anx-A5 to enhance the nucleation and growth of mineral appears to stem from its ability to form two-dimensional crystalline arrays on PS-containing monolayers. However, some stimulatory effect also may result from its ability to exclude Mg(2+) and HCO(-)(3) from nucleation sites. Comparing the various annexins for their ability to activate PS-CPLX nucleation yields the following: avian cartilage Anx-A5 > human placental Anx-A5 > avian liver Anx-A5 > or = avian cartilage Anx-A6 >> cartilage Anx-A2. The stimulatory effect of human placental Anx-A5 and avian cartilage Anx-A6 depended on the presence of PS, since in its absence they either had no effect or actually inhibited the nucleation activity of ACP. Anx-A2 did not significantly enhance mineralization.


Subject(s)
Annexin A5/chemistry , Calcification, Physiologic , Calcium/chemistry , Membranes, Artificial , Models, Biological , Phosphatidylserines/chemistry , Animals , Annexin A2/chemistry , Annexin A2/metabolism , Annexin A5/metabolism , Annexin A6/chemistry , Annexin A6/metabolism , Bicarbonates/chemistry , Bicarbonates/metabolism , Calcium/metabolism , Cations, Divalent/chemistry , Cations, Divalent/metabolism , Chondrocytes/chemistry , Chondrocytes/metabolism , Crystallization , Emulsions/chemistry , Humans , Magnesium/chemistry , Magnesium/metabolism , Phosphatidylserines/metabolism
8.
Anal Biochem ; 367(2): 159-66, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17585866

ABSTRACT

Matrix vesicles (MVs) are involved in de novo mineral formation by nearly all vertebrate tissues. The driving force for MV mineralization is a nucleational core composed of three principal constituents: (i) amorphous calcium phosphate (ACP), complexed in part with phosphatidylserine (PS) to form (ii) calcium-phosphate-lipid complexes (CPLX), and (iii) annexin A5 (AnxA5), the principal lipid-dependent Ca(2+)-binding protein in MVs. We describe methods for reconstituting the nucleational core using a biomimetic approach and for analyzing the kinetics of its induction of mineral formation. The method is based on light scattering by the nascent crystallites at 340 nm and monitors mineral formation at regular intervals without disturbing the system using an automated plate reader. It yields precise replicate values that typically agree within less than 5%. As with MVs, mineral formation by the synthetic complex follows a sigmoidal pattern; following a quiescent induction period, rapid formation ensues for a limited time, followed by a distinct decline in rate that continues to slow, ultimately reaching a maximal asymptotic value. Key to quantization of mineral formation is the use of first-derivative analysis, which defines the induction time, the rate and the amount of initial mineral formation. Furthermore, using a five-parameter logistic curve-fitting algorithm, the maximal amount of mineral formation can be predicted accurately. Using these methods, we document the dramatic finding that AnxA5 synergistically activates PS-CPLX, transforming it from a very weak nucleator of mineral formation to a potent one. The methods presented should enable systematic study of the effects of numerous other factors thought to contribute to mineral formation.


Subject(s)
Annexin A5/physiology , Calcification, Physiologic/physiology , Calcium Phosphates/metabolism , Collagen Type II/physiology , Phosphatidylserines/physiology , Cartilage/physiology , Kinetics , Liposomes/metabolism , Lymph/physiology , Models, Biological
9.
J Cell Physiol ; 202(1): 167-77, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15389532

ABSTRACT

We report here a comparative study of the development and behavior of chondrocytes isolated from normal growth plate tissue, tibial dyschondroplasic lesions, and from articular cartilage. The objective of these studies was to determine whether the properties exhibited by chondrocytes in dysplasic lesions or in articular cartilage were due to their cellular phenotype, their environment, or both. We had previously analyzed the electrolytes and amino acid levels in the extracellular fluid of avian growth plate chondrocytes. Using these data, we constructed a culture medium (DATP5) in which growth plate cells essentially recapitulate their normal behavior in vivo. Here, we used DATP5 to examine the behavior of chondrocytes isolated from lesions of tibial dyschondroplasia (TD). We found that once isolated from lesion and grown in this supportive medium, dysplasic chondrocytes behaved essentially like normal growth plate cells. These findings suggest that the cause of TD is local factors operating in vivo to prevent these cells from developing normally. With respect to articular chondrocytes, our data indicate that they more closely retain normal protein and proteoglycan synthesis when grown in serum-free media. These cells readily induced mineral formation in vitro, both in the presence and absence of serum. However, in serum-containing media, mineralization was significantly enhanced when the cells were exposed to retinoic acid (RA) or osteogenic protein-1 (OP-1). Our studies support previous work indicating the presence of autocrine factors produced by articular chondrocytes in vivo that prevent mineralization and preserve matrix integrity. The lack of inhibitory factors and the presence of supporting factors are likely reasons for the induction of mineralization by articular chondrocytes in vitro.


Subject(s)
Calcification, Physiologic/genetics , Cartilage, Articular/growth & development , Chondrocytes/metabolism , Chondrogenesis/physiology , Growth Plate/growth & development , Osteochondrodysplasias/metabolism , Animals , Autocrine Communication/physiology , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/pharmacology , Calcification, Physiologic/drug effects , Calcinosis/genetics , Calcinosis/metabolism , Calcinosis/physiopathology , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Chickens , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrogenesis/drug effects , Culture Media/chemistry , Culture Media/pharmacology , Culture Media, Serum-Free/pharmacology , Growth Plate/cytology , Growth Plate/drug effects , Growth Plate/metabolism , Osteochondrodysplasias/genetics , Osteochondrodysplasias/physiopathology , Proteoglycans/biosynthesis , Tibia/growth & development , Tibia/pathology , Tibia/physiopathology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Tretinoin/pharmacology
10.
J Cell Biochem ; 88(5): 954-69, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12616534

ABSTRACT

Matrix vesicles released by epiphyseal growth plate chondrocytes are known to contain a significant quantity of labile Zn(2+). Zonal analysis of chicken metatarsal bones showed that the resting/proliferative region of the growth plate contained high levels of Zn(2+) with significantly lower levels in the hypertrophic cartilage suggesting a loss of cellular Zn(2+) as the chondrocytes mature. Intracellular labile Zn(2+) was measured in primary cultures of growth plate chondrocytes by assay with the fluorescent Zn-chelator toluenesulfonamidoquinoline (TSQ) and imaged by multi-photon laser scanning microscopy (MPLSM) with the TSQ derivative zinquin. Short-term exposure to Zn(2+), both in the presence and absence of pyrithione resulted in significant increases in cytosolic Zn(2+). Treatment with the membrane-permeant Zn(2+) chelator TPEN rapidly reduced the levels of labile Zn(2+) and triggered apoptosis. Cytosolic Zn(2+) levels were significantly reduced following 24-h incubations with known inducers of chondrocyte apoptosis. The loss of intracellular Zn(2+) was accompanied by a significant reduction in the cytosolic metal-binding protein metallothionein. Examination of Zn(2+)-treated cells with MPLSM showed uniformly higher zinquin fluorescence. Treatment of Zn(2+)-loaded cells with TPEN quenched zinquin fluorescence confirming that the observed fluorescence in chondrocytes is due to the presence of intracellular Zn(2+). A dose-dependent increase in zinquin fluorescence was observed in cells treated with a range of Zn(2+) concentrations. Short-term treatment of cultured chondrocytes with apoptosis-inducing chemicals resulted in transient increases in intracellular labile Zn(2+). These results indicate that Zn(2+) is mobilized from intracellular binding sites in the early stages of chondrocyte apoptosis and is subsequently lost from the cells. The early mobilization of Zn(2+) provides a mechanism for its movement to matrix vesicles and the extracellular matrix.


Subject(s)
Apoptosis/physiology , Chondrocytes/metabolism , Growth Plate/metabolism , Zinc/metabolism , Animals , Cations, Divalent , Cells, Cultured , Chickens , Chondrocytes/chemistry , Fluorescent Dyes , Growth Plate/chemistry , Metatarsal Bones/growth & development , Microscopy, Confocal , Quinolones , Tibia/growth & development , Tosyl Compounds , Zinc/analysis
11.
J Inorg Biochem ; 94(1-2): 14-27, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12620669

ABSTRACT

Stable, large unilamellar vesicles (LUV) have been constructed that model matrix vesicles (MV) in inducing de novo mineral formation when incubated in synthetic cartilage lymph (SCL). Using a dialysis method for incorporation of predetermined pure lipid, electrolyte and protein constituents, the detergent n-octyl beta-D-glucopyranoside enabled formation of stable, impermeable LUV with a diameter ( approximately 300 nm), lipid composition (phosphatidylcholine-phosphatidylserine-cholesterol, 7:2:2, molar ratio) and enclosed inorganic phosphate level (25-100 mM) similar to that of native MV. Mineral formation by these LUVs was measured by 45Ca(2+) uptake and FTIR analysis following incubation in SCL. Addition of the ionophore A23187 to SCL enabled 45Ca(2+) uptake comparable to that of native MV. FTIR analysis revealed that crystalline mineral formed in the LUV during incubation in SCL, but not in the absence of ionophore. This mineral had an IR absorption spectrum like that of the acid-phosphate-rich, octacalcium phosphate-like mineral formed by native MV. Perturbing the LUV membrane with either detergents or phospholipase A(2) following prior incubation in SCL enabled egress of mineral crystallites from the vesicle lumen, stimulating further mineral formation. Annexin V, a major protein in native MV with known Ca(2+) channel activity, incorporated into the LUV lumen or added to the external medium, induced only limited 45Ca(2+) uptake. This indicates that additional factors are required for annexin V to form Ca(2+) channels. Nevertheless for the first time, stable LUVs have been constructed with MV-like lipid, electrolyte, and protein composition and size that induce formation of mineral like that formed by native MV.


Subject(s)
Models, Biological , Annexin A5/metabolism , Lipid Metabolism , Spectroscopy, Fourier Transform Infrared
12.
J Inorg Biochem ; 94(3): 221-35, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12628702

ABSTRACT

The mechanism of matrix vesicle (MV) mineralization was studied using MVs isolated from normal growth plate tissue, as well as several putative intermediates in the MV mineralization pathway--amorphous calcium phosphate (ACP), calcium phosphate phosphatidylserine complex (CPLX) and hydroxyapatite (HAP). Radionuclide uptake and increase in turbidity were used to monitor mineral formation during incubation in synthetic cartilage lymph (SCL). Inhibitors of phosphate (Pi) metabolism, as well as replacing Na(+) with various cations, were used to study MV Pi transport, which had been thought to be Na(+)-dependent. MVs induced rapid mineralization approximately 3 h after addition to SCL; CPLX and HAP caused almost immediate induction; ACP required approximately 1 h. Phosphonoformate (PFA), a Pi analog, potently delayed the onset and reduced the rate of mineral formation of MV and the intermediates with IC(50)'s of 3-6 microM and approximately 10 microM, respectively. PFA:Pi molar ratios required to reduce the rate of rapid mineralization by 50% were approximately 1:30 for ACP, approximately 1:20 for HAP, approximately 1:3.3 for CPLX, and approximately 1:2.0 for MVs. MV mineralization was not found to be strictly Na(+)-dependent: substitution of Li(+) or K(+) for Na(+) had minimal effect; while N-methyl D-glucamine (NMG(+)) was totally inhibitory, choline(+) was clearly stimulatory. Na(+) substitutions had minimal effect on HAP- and CPLX-seeded mineral formation. However with ACP, NMG(+) totally blocked and choline(+) stimulated, just as they did MV mineralization. Thus, kinetic analyses indicate that ACP is a key intermediate, nevertheless, formation of CPLX appears to be the rate-limiting factor in MV mineralization.


Subject(s)
Calcification, Physiologic/drug effects , Phosphates/pharmacology , Sodium/pharmacology , Animals , Biological Transport, Active , Bone Matrix/drug effects , Bone Matrix/metabolism , Calcium Phosphates/metabolism , Calcium Phosphates/pharmacology , Chickens , Durapatite/metabolism , Durapatite/pharmacology , Growth Plate/drug effects , Growth Plate/growth & development , Growth Plate/metabolism , In Vitro Techniques , Kinetics , Phosphates/metabolism , Phosphatidylserines/metabolism , Phosphatidylserines/pharmacology
13.
Anal Biochem ; 322(1): 104-15, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14705786

ABSTRACT

Matrix vesicles (MV) are lipid bilayer-enclosed nanoscale structures that initiate extracellular mineral formation in most vertebrate species. Little attention has been given to differences between species in membrane lipid composition or to how new mineral is formed in MV. To explore more precisely the lipids of MV isolated from avian and bovine species, we developed a new high-performance liquid chromatography (HPLC) method used in combination with evaporative light scattering detection (ELSD) to quantify their lipid composition. HPLC analyses were performed on a Lichrosorb silica column using separate binary gradient elution systems for analyzing polar and nonpolar lipids. Standard mixtures of both phospholipids and nonpolar lipids were used to prepare calibration curves for each lipid, which were analyzed mathematically by polynomial regression for accurate quantitation. This new methodology provides high-resolution separations and quantitation of both the polar and the nonpolar lipids typically present in biological membranes, including lyso- (monoacyl-) phospholipids. We have applied this method to quantitate the phospholipid and nonpolar lipid composition of MV isolated from chicken and bovine growth plate cartilage. We also compared the ability of these MV to induce mineral formation. While the ability to induce mineralization and the lipid composition were generally similar, some significant differences between MV from these two disparate species were seen.


Subject(s)
Calcification, Physiologic/physiology , Cartilage/metabolism , Growth Plate/metabolism , Lipid Bilayers/analysis , Phospholipids/analysis , Animals , Cattle , Chickens , Chromatography, High Pressure Liquid
14.
J Cell Biochem ; 87(2): 173-87, 2002.
Article in English | MEDLINE | ID: mdl-12244570

ABSTRACT

Sonic hedgehog (Shh) is a key signal protein in early embryological patterning of limb bud development. Its analog, Indian hedgehog (Ihh), primarily expressed during early cartilage development in prehypertrophic chondrocytes, regulates proliferation and suppresses terminal differentiation of postnatal growth plate (GP) chondrocytes. We report here for the first time that both Shh and Ihh mRNA are expressed in the GP of rapidly growing 6-week-old broiler-strain chickens. They are also expressed in other tissues such as articular chondrocytes, kidney, and bone. In situ hybridization and RT-PCR analyses reveal Shh in all zones of the GP, with peak expression in late hypertrophy. Using primary cultures of GP chondrocytes in serum-containing medium, we followed the patterns of Shh and Ihh mRNA expression as the cultures matured and mineralized. We find a cyclical expression of both hedgehog genes during the early period of culture development between day 10 and 14; when one is elevated, the other tended to be suppressed, suggesting that the two hedgehogs may play complementary roles during GP development. Retinoic acid (RA), a powerful modulator of gene expression in cell differentiation, stimulates GP chondrocytes toward terminal differentiation, enhancing mineral formation. We find that RA strongly suppresses Ihh, but enhances expression of Shh in this system. While Ihh suppresses maturation of GP chondrocytes to hypertrophy, we hypothesize that Shh acts to push these cells toward hypertrophy.


Subject(s)
Chondrocytes/metabolism , Growth Plate/metabolism , Trans-Activators/biosynthesis , Tretinoin/pharmacology , Animals , Base Sequence , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Chickens , Chondrocytes/drug effects , Collagen/biosynthesis , Gene Expression Regulation, Developmental , Growth Plate/cytology , Growth Plate/drug effects , Hedgehog Proteins , In Situ Hybridization , Molecular Sequence Data , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Trans-Activators/genetics
15.
J Cell Biochem ; 86(3): 475-89, 2002.
Article in English | MEDLINE | ID: mdl-12210754

ABSTRACT

This report describes Pi transport activity in chondrocytes isolated from the growth plate (GP) of normal adolescent chickens grown in primary cell culture. Our recent work showed that Pi transport in matrix vesicles (MV) isolated from normal GP cartilage was not strictly Na+-dependent, whereas previously characterized Pi transport from rachitic GP cartilage MV was. This Na+-dependent Pi transporter (NaPiT), a member of the Type III Glvr-1 gene family, is expressed only transiently during early differentiation of GP cartilage, is enhanced by Pi-deficiency, and is most active at pH 6.8. Since GP mineralization requires abundant Pi and occurs under slightly alkaline conditions, it seemed unlikely that this type of Pi transporter was solely responsible for Pi uptake during normal GP development. Therefore we asked whether the lack of strict Na+-dependency in Pi transport seen in normal MV was also evident in normal GP chondrocytes. In fact, cellular Pi transport was found not to be strictly Na+-dependent, except for a brief period early in the culture. Choline could equally serve as a Na+ substitute. Activity of choline-supported Pi transport was optimum at pH 7.6-8.0. In addition, prior exposure of the cells to elevated extracellular Pi (2-3 mM) strongly enhanced subsequent Pi uptake, which appeared to depend on prior loading of the cells with mineral ions. Prevention of Pi loading by pretreatment with Pi transport inhibitors not only inhibited subsequent cellular Pi uptake, it also blocked mineral formation. Treatment with elevated extracellular Pi did not induce apoptosis in these GP chondrocytes.


Subject(s)
Chondrocytes/cytology , Chondrocytes/metabolism , Growth Plate/cytology , Phosphates/metabolism , Tibia/cytology , Alendronate/pharmacology , Animals , Biological Transport , Cell Size/drug effects , Cells, Cultured , Chickens , Chondrocytes/drug effects , Culture Media/metabolism , Hydrogen-Ion Concentration , Sexual Maturation , Sodium , Time Factors
16.
J Biol Chem ; 277(7): 5126-33, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11714705

ABSTRACT

Matrix vesicles are lipid bilayer-enclosed structures that initiate extracellular mineral formation. Little attention has been given to how newly formed mineral interacts with the lipid constituents and then emerges from the lumen. To explore whether specific lipids bind to the incipient mineral and if breakdown of the membrane is involved, we analyzed changes in lipid composition and extractability during vesicle-induced calcification. Isolated matrix vesicles were incubated in synthetic cartilage lymph to induce mineral formation. At various times, samples of the lipids were taken for analysis, extracted both before and after demineralization to remove deposited mineral. Phosphatidylserine and phosphatidylinositol both rapidly disappeared from extracts made before decalcification, indicating rapid degradation. However, extracts made after demineralization revealed that phosphatidylserine had become complexed with newly forming mineral. Concomitantly, its levels actually increased, apparently by base-exchange with phosphatidylethanolamine. Though partially complexed with the mineral, phosphatidylinositol was nevertheless rapidly broken down. Sphingomyelin and phosphatidylethanolamine also underwent rapid breakdown, but phosphatidylcholine was degraded more slowly, all accompanied by a buildup of free fatty acids. The data indicate that phosphatidylserine forms complexes that accompany mineral formation, while degradation of other membrane phospholipids apparently enables egress of crystalline mineral from the vesicle lumen.


Subject(s)
Cartilage/metabolism , Growth Plate/metabolism , Lipids/chemistry , Lysophospholipids/chemistry , Phospholipids/metabolism , Animals , Chickens , Chromatography, High Pressure Liquid , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Sphingomyelins/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...