Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Medicina (Kaunas) ; 56(2)2020 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-31972958

RESUMO

Background and Objectives: Maxillary bone defects related to post-extraction alveolar ridge resorption are usual. These defects may lead to failure in further surgical implant phases given the lack of bone volume to perform the dental implant. The objective of this clinical assay was to evaluate the safety and efficacy of an experimental synthetic bone substitute in the preservation of post-extraction maxillary alveoli. Materials and Methods: 33 voluntary patients who had at least one maxillary premolar tooth that was a candidate for exodontia (n = 39) and subsequent implant rehabilitation participated. The regenerated alveoli were monitored by means of periodic clinical examinations (days 9 ± 1, 21 ± 4, 42 ± 6, and 84 ± 6), measuring the height and width of the alveolar crest (days 0 and 180 ± 5), measurement of radiodensity using tomographic techniques (days 0-5 and 175 ± 5), and histological examination of biopsies collected at 180 ± 5 days. Results: No significant differences were observed during the entire follow-up period between the two groups with respect to the safety variables studied. A variation in width of -0.9 ± 1.3 mm and -0.6 ± 1.5 mm, and a variation in height of -0.1 ± 0.9 mm and -0.3 ± 0.7 mm was observed for experimental material Sil-Oss® and Bio-Oss®, respectively. The radiodensity of the alveoli regenerated with the experimental material was significantly lower than that corresponding to Bio-Oss®. However, the histological study showed greater osteoid matrix and replacement of the material with newformed bone in the implanted beds with the experimental material. Conclusions: Both materials can be used safely and proved equally effective in maintaining alveolar flange dimensions, they are also histologically biocompatible, bioactive and osteoconductive. The experimental material showed the advantage of being resorbable and replaced with newformed bone, in addition to promoting bone regeneration.


Assuntos
Perda do Osso Alveolar/tratamento farmacológico , Fosfatos de Cálcio/farmacologia , Durapatita/antagonistas & inibidores , Sílica Gel/farmacologia , Adulto , Perda do Osso Alveolar/prevenção & controle , Substitutos Ósseos/normas , Substitutos Ósseos/uso terapêutico , Fosfatos de Cálcio/uso terapêutico , Método Duplo-Cego , Combinação de Medicamentos , Durapatita/farmacologia , Durapatita/uso terapêutico , Feminino , Humanos , Masculino , Maxila/efeitos dos fármacos , Maxila/fisiopatologia , Pessoa de Meia-Idade , Sílica Gel/uso terapêutico
3.
Calcif Tissue Int ; 99(5): 525-534, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27461215

RESUMO

Vascular calcification significantly contributes to mortality in chronic kidney disease (CKD) patients. Sevelamer and pyrophosphate (PPi) have proven to be effective in preventing vascular calcification, the former by controlling intestinal phosphate absorption, the latter by directly interfering with the hydroxyapatite crystal formation. Since most patients present with established vascular calcification, it is important to evaluate whether these compounds may also halt or reverse the progression of preexisting vascular calcification. CKD and vascular calcification were induced in male Wistar rats by a 0.75 % adenine low protein diet for 4 weeks. Treatment with PPi (30 or 120 µmol/kg/day), sevelamer carbonate (1500 mg/kg/day) or vehicle was started at the time point at which vascular calcification was present and continued for 3 weeks. Hyperphosphatemia and vascular calcification developed prior to treatment. A significant progression of aortic calcification in vehicle-treated rats with CKD was observed over the final 3-week period. Sevelamer treatment significantly reduced further progression of aortic calcification as compared to the vehicle control. No such an effect was seen for either PPi dose. Sevelamer but not PPi treatment resulted in an increase in both osteoblast and osteoid perimeter. Our study shows that sevelamer was able to reduce the progression of moderate to severe preexisting aortic calcification in a CKD rat model. Higher doses of PPi may be required to induce a similar reduction of severe established arterial calcification in this CKD model.


Assuntos
Difosfatos/farmacologia , Durapatita/antagonistas & inibidores , Insuficiência Renal Crônica/complicações , Sevelamer/farmacologia , Calcificação Vascular/patologia , Animais , Aorta/patologia , Quelantes/farmacologia , Masculino , Ratos , Ratos Wistar , Calcificação Vascular/etiologia
4.
Acc Chem Res ; 46(9): 2136-44, 2013 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-23932180

RESUMO

Nature has evolved sophisticated strategies for engineering hard tissues through the interaction of proteins, and ultimately cells, with inorganic mineral phases. This process, called biomineralization, is how living organisms transform inorganic materials such as hydroxyapatite, calcite, and silica into highly intricate and organized structures. The remarkable material properties of shell, bone, and teeth come from the activities of proteins that function at the organic-inorganic interface. A better understanding of the biomolecular mechanisms used to promote or retard the formation of mineral-based structures could provide important design principles for the development of calcification inhibitors and promoters in orthopedics, cardiology, urology, and dentistry. With the knowledge of the structural basis for control of hard tissue growth by proteins, scientists could potentially develop materials using biomimetic principles with applications in catalysis, biosensors, electronic devices, and chromatographic separations, to name a few. Additionally, biomineralization also has potential applications in electronics, catalysis, magnetism, sensory devices, and mechanical design. Where man-made hard materials require the use of extreme temperatures, high pressure, and pH, biological organisms can accomplish these feats at ambient temperature and at physiological pH. Despite the fact that many researchers want to identify and control the structure of proteins at material and biomineral interfaces, there is a decided lack of molecular-level structure information available for proteins at biomaterial interfaces in general. In particular, this holds for mammalian proteins that directly control calcification processes in hard tissue. The most fundamental questions regarding the secondary and tertiary structures of proteins adsorbed to material surfaces, how proteins catalyze the formation of biomineral composites, or how proteins interact at biomaterial interfaces remain unanswered. This is largely due to a lack of methods capable of providing high-resolution structural information for proteins adsorbed to material surfaces under physiologically relevant conditions. In this Account, we highlight recent work that is providing insight into the structure and crystal recognition mechanisms of a salivary protein model system, as well as the structure and interactions of a peptide that catalyzes the formation of biosilica composites. To develop a better understanding of the structure and interactions of proteins in biomaterials, we have used solid-state NMR techniques to determine the molecular structure and dynamics of proteins and peptides adsorbed onto inorganic crystal surfaces and embedded within biomineral composites. This work adds to the understanding of the structure and crystal recognition mechanisms of an acidic human salivary phosphoprotein, statherin.


Assuntos
Espectroscopia de Ressonância Magnética , Peptídeos/química , Proteínas/química , Carbonato de Cálcio/química , Durapatita/antagonistas & inibidores , Durapatita/química , Humanos , Microscopia Eletrônica de Varredura , Modelos Moleculares , Proteínas e Peptídeos Salivares/química , Dióxido de Silício/química
5.
J Trace Elem Med Biol ; 27(4): 346-51, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23867705

RESUMO

PROJECT: Aluminum (Al) is an increasing problem in biomedicine since it can interact with phosphates. Bone is one of the preferential target tissues of Al deposition: Al interacts with mineralization and/or bone cell activities. We searched the influence of Al deposition in hydroxyapatite developed on a biomimetic polymer (carboxymethylated poly(2-hydroxyethyl-methacrylate)) which mimics bone mineralization in the absence of cells. PROCEDURES: Pellets of polymer were incubated for 5 days in a synthetic body fluid (SBF) to induce mineralization, then 21 days in SBF containing 20, 40 and 60 µg/L Al(3+). Other pellets were incubated in SBF containing commercial Al foil (33 mg/vial) either in 1, 2 or 6 pieces. The mineral deposits were dissolved in HCl and Ca(2+), PO(4)(3-) and Al(3+) content was measured. Hydroxyapatite was characterized by SEM and X energy-dispersive X-ray analysis (EDX). RESULTS: The amount of Al(3+) was dose-dependently increased in Ca/P deposits on the polymer pellets. At high concentration (or with the 6 Al foils) growth of hydroxyapatite calcospherite was inhibited; only calcified plates emerging from the polymer were observed. Pellets incubated with 1 and 2 Al foils exhibited a reduction in calcospherite diameter and an increase in the Al(3+)/Ca(2+) ratio. EDX identified Al in the mineral deposits. CONCLUSIONS: In this acellular model, Al(3+) altered the growth of calcospherites at low concentration and inhibited their development at high concentration. In SBF, a release of Al(3+) from aluminum foils also inhibited mineralization. This study emphasizes the importance of Al in bone pathology and stresses the question of its release from biomaterials.


Assuntos
Alumínio/farmacologia , Materiais Biomiméticos/química , Durapatita/antagonistas & inibidores , Poli-Hidroxietil Metacrilato/química , Alumínio/química , Materiais Biomiméticos/síntese química , Cristalização , Durapatita/síntese química , Durapatita/química , Tamanho da Partícula , Poli-Hidroxietil Metacrilato/síntese química , Propriedades de Superfície
6.
Curr Med Chem ; 18(14): 2196-203, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21517761

RESUMO

In this review we consider diseases associated with pathological mineralization/ossification, namely, ankylosing spondylitis (AS), osteoarthritis (OA), generalized artery calcification of infancy (GACI), vascular calcification as well as chondrocalcinosis (CC) and pseudo gout. Deciphering the key enzymes implicated in the calcification process is an objective of prime importance and the ultimate goal is to synthesize inhibitors of these enzymes in order to provide efficient alternate therapeutic strategies that will slow down the pathologic mineralization and complement the arsenal of anti-inflammatory drugs. One of the difficulties in the definition of diseases associated with pathologic mineralization/ossification lies in the controversial relationship between the type of calcification and the nature of the disease. Here, we propose to clarify this relationship by making a distinction between diseases associated with hydroxyapatite (HA) and calcium pyrophosphate dihydrate (CPPD) deposits. AS, OA, GACI and vascular calcification are usually characterized by mineralization/ossification associated with HA deposits, while CC and pseudo gout are mostly characterized by CPPD deposits. Although both HA and CPPD deposits may occur concomitantly, as in chronic pyrophosphate arthritis or in OA with CPPD, they are formed as a result of two antagonistic processes indicating that treatment of distinct diseases can be only achieved by disease-specific drug therapies. The hydrolysis of PPi, an inhibitor of HA formation, is mostly controlled by tissue non-specific alkaline phosphatase TNAP, while PPi production in the extracellular medium is controlled by ANK, a PPi transporter, and/or NPP1 which generates PPi from nucleotide triphosphates. Low PPi concentration may lead to a preferential deposition of HA while high PPi concentration will favor the formation of CPPD deposits. Thus, HA and CCPD deposition cannot occur concomitantly because they are determined by the Pi/PPi ratio which, in turn, depends on the relative activities of antagonistic enzymes, TNAP hydrolyzing PPi or ANK and NPP1 producing PPi. TNAP inhibitors could prevent HA formation in AS, in late OA, in GACI, as well as in vascular calcifications, while ANK or NPP1 inhibitors could slow down CCPD deposition in CC and pseudo gout.


Assuntos
Calcinose/metabolismo , Pirofosfato de Cálcio/metabolismo , Condrocalcinose/metabolismo , Durapatita/metabolismo , Osteoartrite/metabolismo , Espondilite Anquilosante/metabolismo , Doenças Vasculares/metabolismo , Fosfatase Alcalina/antagonistas & inibidores , Fosfatase Alcalina/metabolismo , Animais , Calcinose/tratamento farmacológico , Calcinose/enzimologia , Pirofosfato de Cálcio/antagonistas & inibidores , Condrocalcinose/tratamento farmacológico , Condrocalcinose/enzimologia , Durapatita/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Osteoartrite/tratamento farmacológico , Osteoartrite/enzimologia , Espondilite Anquilosante/tratamento farmacológico , Espondilite Anquilosante/enzimologia , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/enzimologia
7.
Nat Mater ; 9(12): 1004-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20972429

RESUMO

Bone is a composite material in which collagen fibrils form a scaffold for a highly organized arrangement of uniaxially oriented apatite crystals. In the periodic 67 nm cross-striated pattern of the collagen fibril, the less dense 40-nm-long gap zone has been implicated as the place where apatite crystals nucleate from an amorphous phase, and subsequently grow. This process is believed to be directed by highly acidic non-collagenous proteins; however, the role of the collagen matrix during bone apatite mineralization remains unknown. Here, combining nanometre-scale resolution cryogenic transmission electron microscopy and cryogenic electron tomography with molecular modelling, we show that collagen functions in synergy with inhibitors of hydroxyapatite nucleation to actively control mineralization. The positive net charge close to the C-terminal end of the collagen molecules promotes the infiltration of the fibrils with amorphous calcium phosphate (ACP). Furthermore, the clusters of charged amino acids, both in gap and overlap regions, form nucleation sites controlling the conversion of ACP into a parallel array of oriented apatite crystals. We developed a model describing the mechanisms through which the structure, supramolecular assembly and charge distribution of collagen can control mineralization in the presence of inhibitors of hydroxyapatite nucleation.


Assuntos
Apatitas/metabolismo , Colágeno Tipo I/metabolismo , Durapatita/antagonistas & inibidores , Animais , Osso e Ossos/metabolismo , Fosfatos de Cálcio/metabolismo , Colágeno Tipo I/química , Microscopia Crioeletrônica , Criopreservação , Tomografia com Microscopia Eletrônica , Matriz Extracelular/metabolismo , Cavalos , Luz , Modelos Moleculares , Nanopartículas/química , Osteogênese , Peptídeos/metabolismo , Espalhamento de Radiação , Espectrometria por Raios X , Coloração e Rotulagem/métodos , Propriedades de Superfície , Tendões/química
9.
Langmuir ; 26(12): 9899-904, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20491496

RESUMO

The effects of three acidic hexapeptides on in vitro hydroxyapatite growth were characterized by pH-stat kinetic studies, adsorption isotherms, and molecular modeling. The three peptides, pSDEpSDE, SDESDE, and DDDDDD, are equal-length model compounds for the acidic sequences in osteopontin, a protein that inhibits mineral formation in both calcified and noncalcified tissues. Growth rates from 1.67 mM calcium and 1.00 mM phosphate solution were measured at pH 7.4 and 37 degrees C in 150 mM NaCl. pSDEpSDE was a strong growth inhibitor when preadsorbed onto hydroxyapatite (HA) seeds from > or = 0.67 mM solutions, concentrations where adsorption isotherms showed relatively complete surface coverage. The nonphosphorylated SDESDE control showed no growth inhibition. Although it adsorbed to almost the same extent as pSDEpSDE, it rapidly desorbed under the pH-stat growth conditions while pSDEpSDE did not. DDDDDD exhibited weak inhibition as its concentration was increased and similar adsorption/desorption behavior to pSDEpSDE. Molecular modeling yielded binding energy trends based on simple adsorption of peptides on the [100] surface that were consistent with observed inhibition, but not for the [001] surface. The relatively unfavorable binding energies for peptides on the [001] surface suggest that their absorption will be primarily on the [100] face. The kinetic and adsorption data are consistent with phosphorylation of osteopontin acting to control mineral formation.


Assuntos
Durapatita/antagonistas & inibidores , Osteopontina/química , Fragmentos de Peptídeos/farmacologia , Adsorção , Animais , Calcificação Fisiológica , Durapatita/química , Humanos , Concentração de Íons de Hidrogênio , Cinética , Modelos Moleculares , Fosforilação
10.
J Periodontal Res ; 44(5): 616-21, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18973518

RESUMO

BACKGROUND AND OBJECTIVE: Dental calculus occurs as a consequence of supersaturation of saliva with respect to calcium phosphates. This mineralization of dental plaque can be delayed by the presence of crystallization inhibitors, such as pyrophosphate or bisphosphonates. Phytate inhibits brushite and hydroxyapatite crystallization and has the potential to prevent dental calculi formation. The aim of the present study was to examine the effects of phytate and zinc, administered in a mouthwash solution, to prevent the formation of dental calculus. MATERIAL AND METHODS: Healthy dental plaque-forming volunteers (n = 25) took part in a randomized, double-blind, three-period crossover clinical study to assess the efficacy of a phytate-containing mouthwash in relation to control and placebo effects. Subjects rinsed their mouths for 1 min, twice each day, with 20 mL of the test solution, without ingestion. Mouthwash efficacy was assessed through quantification of the amounts of calcium, phosphorus and magnesium present in the residues obtained by dental cleaning, performed by a single trained examiner. RESULTS: A good correlation was found among total calcium, magnesium and phosphorus in calcified dental plaque residues, indicating that any of these variables is adequate for evaluating the reduction of plaque crystallization as calcium phosphate. A statistically significant decrease in total calcium, magnesium and phosphorus was found in the phytate-treatment period compared with control and placebo periods, demonstrating the efficacy of the proposed treatment in reducing dental calculus formation. CONCLUSION: The high efficacy of phytate in reducing dental calculus formation suggests that this substance may be an effective treatment for preventing the development of calculus deposits.


Assuntos
Anti-Infecciosos Locais/uso terapêutico , Cálculos Dentários/prevenção & controle , Antissépticos Bucais/uso terapêutico , Ácido Fítico/uso terapêutico , Triclosan/uso terapêutico , Adolescente , Adulto , Idoso , Cálcio/análise , Fosfatos de Cálcio/antagonistas & inibidores , Estudos Cross-Over , Cristalização , Placa Dentária/química , Método Duplo-Cego , Durapatita/antagonistas & inibidores , Feminino , Humanos , Magnésio/análise , Masculino , Pessoa de Meia-Idade , Fósforo/análise , Placebos , Adulto Jovem , Zinco/uso terapêutico
11.
Ann Thorac Surg ; 79(3): 1072-80, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15734452

RESUMO

Calcification plays a major role in the failure of bioprosthetic and other tissue heart valve substitutes. Tissue valve calcification is initiated primarily within residual cells that have been devitalized, usually by glutaraldehyde pretreatment. The mechanism involves reaction of calcium-containing extracellular fluid with membrane-associated phosphorus to yield calcium phosphate mineral deposits. Calcification is accelerated by young recipient age, valve factors such as glutaraldehyde fixation, and increased mechanical stress. Recent studies have suggested that pathologic calcification is regulated by inductive and inhibitory factors, similar to the physiologic mineralization of bone. The most promising preventive strategies have included binding of calcification inhibitors to glutaraldehyde fixed tissue, removal or modification of calcifiable components, modification of glutaraldehyde fixation, and use of tissue cross linking agents other than glutaraldehyde. This review summarizes current concepts in the pathophysiology of tissue valve calcification, including emerging concepts of endogenous regulation, progress toward prevention of calcification, and issues related to calcification of the aortic wall of stentless bioprosthetic valves.


Assuntos
Bioprótese/efeitos adversos , Calcinose/etiologia , Calcinose/prevenção & controle , Glutaral/efeitos adversos , Próteses Valvulares Cardíacas/efeitos adversos , Animais , Durapatita/antagonistas & inibidores , Humanos
12.
J Periodontol ; 75(4): 537-45, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15152817

RESUMO

BACKGROUND: A newly developed bisphosphonate, TRK-530 (disodium dihydrogen[4-(methylthio)phenylthio]methanebisphosphonate), has recently been reported to show anti-inflammatory and anti-bone-resorbing activity. Since bisphosphonates have been shown to inhibit the formation of calcium-phosphate crystals in vitro, TRK-530 may inhibit the formation of dental calculus. Therefore, the present study was performed to examine whether this compound has such an effect. METHODS: Three groups of Wistar rats fed a calculogenic diet (RC16) were treated with TRK-530 in drinking water at concentrations of 0 (control group), 0.75, and 1.5 mM. Another group received a daily subcutaneous injection of TRK-530 at a dose of 2.25 micromoles/rat, which was assumed to correspond to the maximum amount of this compound absorbed from the intestine when rats received 1.5 mM TRK-530 in drinking water. Rat dental calculus formation was evaluated. The crystalline nature of dental calculus was studied by x-ray diffraction analysis. Finally, the effects of TRK-530 on the precipitation of calcium-phosphate from solution were tested in vitro. RESULTS: TRK-530 in drinking water inhibited dental calculus formation dose-dependently. However, subcutaneous injection of TRK-530 did not have any significant effect, suggesting that the anticalculus effect of TRK-530 in drinking water was topical, not systemic. The calculus that formed in both the control and experimental groups was primarily hydroxyapatite, a main constituent of human dental calculus. TRK-530 inhibited the precipitation of calcium-phosphate from solution in vitro. CONCLUSIONS: TRK-530 inhibited the formation of dental calculus in a dose-dependent fashion via a local effect. Inhibition of the precipitation of calcium-phosphate from solution might be involved in the anticalculogenic mechanism of this drug.


Assuntos
Cálculos Dentários/prevenção & controle , Difosfonatos/administração & dosagem , Administração Tópica , Análise de Variância , Animais , Precipitação Química , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Ingestão de Líquidos , Durapatita/antagonistas & inibidores , Masculino , Ratos , Ratos Wistar , Água
13.
Arch Oral Biol ; 42(8): 527-37, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9347115

RESUMO

Phosphoproteins in human saliva include proline-rich proteins, statherins, histatin 1 and cystatin SA-III. The presence of phosphate in these proteins is necessary for various functions in the mouth including calcium binding, inhibition of precipitation of calcium phosphate, inhibition of growth of hydroxyapatite crystals and adherence to hydroxyapatite. To elucidate the process of phosphorylation of these proteins, the phosphorylation of a peptide (APRP8) with an amino acid sequence identical to one of the phosphorylated sites in acidic proline-rich proteins by a kinase from the human sublingual gland was investigated. The kinase, which was highly labile, was purified 58-fold by fractionation of sublingual gland homogenate and gel filtration, but the enzyme was inactivated when further purification by chromatographic techniques commonly used for protein kinases was attempted. To compare the enzyme with other kinases, and to obtain information that could be used in its further purification, a characterization was undertaken. The enzyme required 10 mM Mg2+ for optimum activity, it had a KM of 0.09 mM for ATP and the KM for the peptide substrate APRP8 was 0.42 mM. It was not activated by cAMP or calmodulin, characteristics that are shared with casein kinases and mammary gland kinase. The sublingual kinase as well as casein kinase 2 were inhibited by heparin, but in other respects the two kinases had different properties. While casein kinase 2 is activated by polylysine and has optimal activity in 150 mM KCl, sublingual kinase was inhibited by polylysine and the addition of KCl. Moreover, casein kinase 2 can utilize both ATP and GTP as phosphoryl donors, but GTP was not a substrate for sublingual kinase. The sublingual kinase shared a substrate recognition sequence with mammary gland kinase, but, unlike that kinase, it could not utilize Ca2+ instead of Mg2+. While the sublingual kinase thus shared some properties with both casein kinase 2 and mammary gland kinase, distinct differences were also seen and the relationship to these enzymes remains to be determined. The characterization of the sublingual kinase will be useful in its further purification.


Assuntos
Fosfoproteínas/metabolismo , Proteínas Quinases/metabolismo , Proteínas e Peptídeos Salivares/metabolismo , Glândula Sublingual/enzimologia , Trifosfato de Adenosina/metabolismo , Adesividade , Mama/enzimologia , Cálcio/metabolismo , Fosfatos de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Caseína Quinases , Precipitação Química , Cromatografia , Cromatografia em Gel , Cistatinas/metabolismo , Inibidores de Cisteína Proteinase/metabolismo , Durapatita/antagonistas & inibidores , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Glicoproteínas/metabolismo , Guanosina Trifosfato/metabolismo , Heparina/farmacologia , Histatinas , Humanos , Isoenzimas/antagonistas & inibidores , Magnésio/metabolismo , Peptídeos/metabolismo , Fosfopeptídeos/metabolismo , Fosforilação , Polilisina/farmacologia , Domínios Proteicos Ricos em Prolina , Inibidores de Proteínas Quinases , Cistatinas Salivares , Glândula Sublingual/metabolismo
14.
Am J Physiol ; 273(2 Pt 2): F224-33, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9277583

RESUMO

Adhesion of microcrystals that nucleate in tubular fluid to the apical surface of renal tubular cells could be a critical step in the formation of kidney stones, 20% of which contain hydroxyapatite (HA). HA crystals bound rapidly to monolayer cultures of monkey kidney epithelial cells (BSC-1 line), used to model the surface of the nephron, in a concentration-dependent manner. Adhesion was blocked by diverse polyanions including heparin, pentosan polysulfate, polyaspartate, and polyglutamate, as well as many found in tubular fluid such as chondroitin sulfates A and B, heparan sulfate, citrate, nephrocalcin, and osteopontin. The polycations cetylpyridinium chloride and cationized ferritin, as well as the cationic dyes alcian blue, polyethylenimine, and brilliant blue R, also inhibited adhesion of HA crystals, as did specific lectins including Triticum vulgaris (wheat germ agglutinin). Anions that inhibited adhesion of crystals appeared to act on the crystal surface, whereas cations and lectins exerted their effect on the cell. Treatment of cells with neuraminidase inhibited binding of crystals, suggesting that anionic cell surface sialic acid residues function as HA crystal receptor sites that can be blocked by specific cations or lectins. Adherence of HA crystals to cells of another renal line (MDCK) and, to 3T3 fibroblasts was also inhibited by heparin, polyaspartate, alcian blue, and T vulgaris lectin, suggesting that these crystals bind to analogous molecules on the surface of different types of cells. These results suggests that the structure, quantity, and/or function of soluble anions in tubular fluid, as well as those anchored to the cell surface, could be critical determinants of HA crystal retention in the nephron and the subsequent formation of a renal stone.


Assuntos
Ânions/metabolismo , Durapatita/metabolismo , Rim/metabolismo , Animais , Ânions/farmacologia , Sítios de Ligação , Cátions/farmacologia , Membrana Celular/metabolismo , Chlorocebus aethiops , Cristalização , Cães , Durapatita/antagonistas & inibidores , Enzimas/farmacologia , Células Epiteliais , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Rim/citologia , Rim/efeitos dos fármacos , Lectinas/farmacologia , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...