Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(21)2022 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-36361965

RESUMEN

Mineralization-competent cells like osteoblasts and chondrocytes release matrix vesicles (MVs) which accumulate Ca2+ and Pi, creating an optimal environment for apatite formation. The mineralization process requires the involvement of proteins, such as annexins (Anx) and tissue-nonspecific alkaline phosphatase (TNAP), as well as low molecular-weight compounds. Apigenin, a flavonoid compound, has been reported to affect bone metabolism, but there are doubts about its mechanism of action under physiological and pathological conditions. In this report, apigenin potency to modulate annexin A6 (AnxA6)- and TNAP-mediated osteoblast mineralization was explored using three cell lines: human fetal osteoblastic hFOB 1.19, human osteosarcoma Saos-2, and human coronary artery smooth muscle cells HCASMC. We compared the mineralization competence, the morphology and composition of minerals, and the protein distribution in control and apigenin-treated cells and vesicles. The mineralization ability was monitored by AR-S/CPC analysis, and TNAP activity was determined by ELISA assay. Apigenin affected the mineral structure and modulated TNAP activity depending on the concentration. We also observed increased mineralization in Saos-2 cells. Based on TEM-EDX, we found that apigenin influenced the mineral composition. This flavonoid also disturbed the intracellular distribution of AnxA6 and TNAP, especially blocking AnxA6 aggregation and TNAP attachment to the membrane, as examined by FM analysis of cells and TEM-gold analysis of vesicles. In summary, apigenin modulates the mineralization process by regulating AnxA6 and TNAP, as well as through various effects on normal and cancer bone tissues or atherosclerotic soft tissue.


Asunto(s)
Apigenina , Calcificación Fisiológica , Humanos , Fosfatasa Alcalina/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Anexina A6/efectos de los fármacos , Anexina A6/metabolismo , Apigenina/farmacología , Apigenina/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/fisiología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo
2.
Int J Mol Sci ; 23(16)2022 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-36012211

RESUMEN

The biochemical machinery involved in matrix vesicles-mediated bone mineralization involves a specific set of lipids, enzymes, and proteins. Annexins, among their many functions, have been described as responsible for the formation and stabilization of the matrix vesicles' nucleational core. However, the specific role of each member of the annexin family, especially in the presence of type-I collagen, remains to be clarified. To address this issue, in vitro mineralization was carried out using AnxA6 (in solution or associated to the proteoliposomes) in the presence or in the absence of type-I collagen, incubated with either amorphous calcium phosphate (ACP) or a phosphatidylserine-calcium phosphate complex (PS-CPLX) as nucleators. Proteoliposomes were composed of 1,2-dipalmitoylphosphatidylcholine (DPPC), 1,2-dipalmitoylphosphatidylcholine: 1,2-dipalmitoylphosphatidylserine (DPPC:DPPS), and DPPC:Cholesterol:DPPS to mimic the outer and the inner leaflet of the matrix vesicles membrane as well as to investigate the effect of the membrane fluidity. Kinetic parameters of mineralization were calculated from time-dependent turbidity curves of free Annexin A6 (AnxA6) and AnxA6-containing proteoliposomes dispersed in synthetic cartilage lymph. The chemical composition of the minerals formed was investigated by Fourier transform infrared spectroscopy (FTIR). Free AnxA6 and AnxA6-proteoliposomes in the presence of ACP were not able to propagate mineralization; however, poorly crystalline calcium phosphates were formed in the presence of PS-CPLX, supporting the role of annexin-calcium-phosphatidylserine complex in the formation and stabilization of the matrix vesicles' nucleational core. We found that AnxA6 lacks nucleation propagation capacity when incorporated into liposomes in the presence of PS-CPLX and type-I collagen. This suggests that AnxA6 may interact either with phospholipids, forming a nucleational core, or with type-I collagen, albeit less efficiently, to induce the nucleation process.


Asunto(s)
Anexina A6 , Calcinosis , 1,2-Dipalmitoilfosfatidilcolina/química , Anexina A6/metabolismo , Colágeno/metabolismo , Humanos , Fosfatos/metabolismo , Fosfatidilserinas/química , Proteolípidos
3.
Int J Mol Sci ; 22(8)2021 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-33924370

RESUMEN

The mineralization process is initiated by osteoblasts and chondrocytes during intramembranous and endochondral ossifications, respectively. Both types of cells release matrix vesicles (MVs), which accumulate Pi and Ca2+ and form apatites in their lumen. Tissue non-specific alkaline phosphatase (TNAP), a mineralization marker, is highly enriched in MVs, in which it removes inorganic pyrophosphate (PPi), an inhibitor of apatite formation. MVs then bud from the microvilli of mature osteoblasts or hypertrophic chondrocytes and, thanks to the action of the acto-myosin cortex, become released to the extracellular matrix (ECM), where they bind to collagen fibers and propagate mineral growth. In this report, we compared the mineralization ability of human fetal osteoblastic cell line (hFOB 1.19 cells) with that of osteosarcoma cell line (Saos-2 cells). Both types of cells were able to mineralize in an osteogenic medium containing ascorbic acid and beta glycerophosphate. The composition of calcium and phosphate compounds in cytoplasmic vesicles was distinct from that in extracellular vesicles (mostly MVs) released after collagenase-digestion. Apatites were identified only in MVs derived from Saos-2 cells, while MVs from hFOB 1.19 cells contained amorphous calcium phosphate complexes. In addition, AnxA6 and AnxA2 (nucleators of mineralization) increased mineralization in the sub-membrane region in strongly mineralizing Saos-2 osteosarcoma, where they co-localized with TNAP, whereas in less mineralizing hFOB 1.19 osteoblasts, AnxA6, and AnxA2 co-localizations with TNAP were less visible in the membrane. We also observed a reduction in the level of fetuin-A (FetuA), an inhibitor of mineralization in ECM, following treatment with TNAP and Ca channels inhibitors, especially in osteosarcoma cells. Moreover, a fraction of FetuA was translocated from the cytoplasm towards the plasma membrane during the stimulation of Saos-2 cells, while this displacement was less pronounced in stimulated hFOB 19 cells. In summary, osteosarcoma Saos-2 cells had a better ability to mineralize than osteoblastic hFOB 1.19 cells. The formation of apatites was observed in Saos-2 cells, while only complexes of calcium and phosphate were identified in hFOB 1.19 cells. This was also evidenced by a more pronounced accumulation of AnxA2, AnxA6, FetuA in the plasma membrane, where they were partly co-localized with TNAP in Saos-2 cells, in comparison to hFOB 1.19 cells. This suggests that both activators (AnxA2, AnxA6) and inhibitors (FetuA) of mineralization were recruited to the membrane and co-localized with TNAP to take part in the process of mineralization.


Asunto(s)
Anexina A2/metabolismo , Anexina A6/metabolismo , Calcificación Fisiológica , Osteoblastos/metabolismo , Osteosarcoma/metabolismo , alfa-2-Glicoproteína-HS/metabolismo , Fosfatasa Alcalina/metabolismo , Calcio/metabolismo , Línea Celular Tumoral , Forma de la Célula , Humanos , Fósforo/metabolismo
4.
Int J Mol Sci ; 21(4)2020 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-32085611

RESUMEN

Annexin A6 (AnxA6) is the largest member of the annexin family of proteins present in matrix vesicles (MVs). MVs are a special class of extracellular vesicles that serve as a nucleation site during cartilage, bone, and mantle dentin mineralization. In this study, we assessed the localization of AnxA6 in the MV membrane bilayer using native MVs and MV biomimetics. Biochemical analyses revealed that AnxA6 in MVs can be divided into three distinct groups. The first group corresponds to Ca2+-bound AnxA6 interacting with the inner leaflet of the MV membrane. The second group corresponds to AnxA6 localized on the surface of the outer leaflet. The third group corresponds to AnxA6 inserted in the membrane's hydrophobic bilayer and co-localized with cholesterol (Chol). Using monolayers and proteoliposomes composed of either dipalmitoylphosphatidylcholine (DPPC) to mimic the outer leaflet of the MV membrane bilayer or a 9:1 DPPC:dipalmitoylphosphatidylserine (DPPS) mixture to mimic the inner leaflet, with and without Ca2+, we confirmed that, in agreement with the biochemical data, AnxA6 interacted differently with the MV membrane. Thermodynamic analyses based on the measurement of surface pressure exclusion (πexc), enthalpy (ΔH), and phase transition cooperativity (Δt1/2) showed that AnxA6 interacted with DPPC and 9:1 DPPC:DPPS systems and that this interaction increased in the presence of Chol. The selective recruitment of AnxA6 by Chol was observed in MVs as probed by the addition of methyl-ß-cyclodextrin (MßCD). AnxA6-lipid interaction was also Ca2+-dependent, as evidenced by the increase in πexc in negatively charged 9:1 DPPC:DPPS monolayers and the decrease in ΔH in 9:1 DPPC:DPPS proteoliposomes caused by the addition of AnxA6 in the presence of Ca2+ compared to DPPC zwitterionic bilayers. The interaction of AnxA6 with DPPC and 9:1 DPPC:DPPS systems was distinct even in the absence of Ca2+ as observed by the larger change in Δt1/2 in 9:1 DPPC:DPPS vesicles as compared to DPPC vesicles. Protrusions on the surface of DPPC proteoliposomes observed by atomic force microscopy suggested that oligomeric AnxA6 interacted with the vesicle membrane. Further work is needed to delineate possible functions of AnxA6 at its different localizations and ways of interaction with lipids.


Asunto(s)
Anexina A6/metabolismo , Calcificación Fisiológica , Matriz Extracelular/metabolismo , Vesículas Extracelulares/metabolismo , 1,2-Dipalmitoilfosfatidilcolina/química , Rastreo Diferencial de Calorimetría , Colesterol/metabolismo , Humanos , Membrana Dobles de Lípidos/metabolismo , Microdominios de Membrana/metabolismo , Microscopía de Fuerza Atómica , Proteolípidos/metabolismo
5.
J Cell Biochem ; 120(4): 6580-6588, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30390318

RESUMEN

BACKGROUND: Niemann Pick type C (NPC) lysosomal disorder is linked to the disruption of cholesterol transport. Recent data suggest that the molecular background of this disease is more complex. It was found that accumulation of cholesterol and glycolipids in the late endosomal/lysosomal compartment of NPC1 cells may affect mitochondrial functions. MATERIALS AND METHODS: In this study, primary skin fibroblasts derived from skin biopsies of two anonymous patients with NPC-carrying mutations in the NPC1 gene, characterized by a high total cholesterol content, as well as two healthy donors were used. The presence of signaling proteins in the whole cell lysates and mitochondrial fractions were examined by Western blotting assay. RESULTS: In this report, we provide experimental evidence that in NPC1 cells, dysfunction of mitochondria and cellular metabolism, as reported by Wos et al in 2016, coexist with alterations in signal transduction pathways, such as the mammalian target of rapamycin, AKT, phosphoinositide-dependent protein kinase-1, glycogen synthase kinase-3 ß, and Jun amino-terminal kinase, leading to abnormal cholesterol accumulation and distribution. CONCLUSION: Differences in signal transduction between control and NPC1 cells may suggest that the latter cells experienced significant alterations in the complex molecular mechanisms that control cellular energy metabolism and vesicular transport.


Asunto(s)
Colesterol/metabolismo , Fibroblastos/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Mitocondrias/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Piel/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transporte Biológico , Estudios de Casos y Controles , Metabolismo Energético , Fibroblastos/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mitocondrias/patología , Mutación , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/patología , Piel/patología
6.
Arch Biochem Biophys ; 667: 14-21, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-30998909

RESUMEN

Matrix vesicles (MVs) are a class of extracellular vesicles that initiate mineralization in cartilage, bone, and other vertebrate tissues by accumulating calcium ions (Ca2+) and inorganic phosphate (Pi) within their lumen and forming a nucleation core (NC). After further sequestration of Ca2+ and Pi, the NC transforms into crystalline complexes. Direct evidence of the existence of the NC and its maturation have been provided solely by analyses of dried samples. We isolated MVs from chicken embryo cartilage and used atomic force microscopy peak force quantitative nanomechanical property mapping (AFM-PFQNM) to measure the nanomechanical and morphological properties of individual MVs under both mineralizing (+Ca2+) and non-mineralizing (-Ca2+) fluid conditions. The elastic modulus of MVs significantly increased by 4-fold after incubation in mineralization buffer. From AFM mapping data, we inferred the morphological changes of MVs as mineralization progresses: prior to mineralization, a punctate feature, the NC, is present within MVs and this feature grows and stiffens during mineralization until it occupies most of the MV lumen. Dynamic light scattering showed a significant increase in hydrodynamic diameter and no change in the zeta potential of hydrated MVs after incubation with Ca2+. This validates that crystalline complexes, which are strongly negative relative to MVs, were forming within the lumen of MVs. These data were substantiated by transmission electron microscopy energy dispersive X-ray and Fourier transform infrared spectroscopic analyses of dried MVs, which provide evidence that the complexes increased in size, crystallinity, and Ca/P ratio within MVs during the mineralization process.


Asunto(s)
Biomineralización/fisiología , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Microscopía de Fuerza Atómica/métodos , Animales , Fenómenos Biomecánicos , Cartílago/química , Cartílago/metabolismo , Cartílago/ultraestructura , Embrión de Pollo , Vesículas Extracelulares/ultraestructura , Microscopía Electrónica de Transmisión , Espectroscopía Infrarroja por Transformada de Fourier
7.
BMC Complement Altern Med ; 19(1): 140, 2019 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-31221152

RESUMEN

BACKGROUND: Cholera is one of the most deadly diarrheal diseases that require new treatments. We investigated the neutralization of cholera toxin by five plant extracts obtained from the Rosaceae family that have been traditionally used in Poland to treat diarrhea (of unknown origin). METHODS: Hot water extracts were prepared from the dried plant materials and lyophilized before phytochemical analysis and assessment of antimicrobial activity using microdilution assays. The ability of the plant extracts to neutralize cholera toxin was analyzed by measurement of cAMP levels in cell cultures, enzyme-linked immunosorbent assay and electrophoresis, as well as flow cytometry and fluorescence microscopy studies of fluorescent-labeled cholera toxins with cultured human fibroblasts. RESULTS: The antimicrobial assays displayed modest bacteriostatic potentials. We found that the plant extracts modulate the effects of cholera toxin on intracellular cAMP levels. Three plant extracts (Agrimonia eupatoria L., Rubus fruticosus L., Fragaria vesca L.) suppressed the binding of subunit B of cholera toxin to the cell surface and immobilized ganglioside GM1 while two others (Rubus idaeus L., Rosa.canina L.) interfered with the toxin internalization process. CONCLUSIONS: The traditional application of the Rosaceae plant infusions for diarrhea appears relevant to cholera, slowing the growth of pathogenic bacteria and either inhibiting the binding of cholera toxin to receptors or blocking toxin internalization. The analyzed plant extracts are potential complements to standard antibiotic treatment and Oral Rehydration Therapy for the treatment of cholera.


Asunto(s)
Antibacterianos/farmacología , Toxina del Cólera/toxicidad , Cólera/microbiología , Extractos Vegetales/farmacología , Rosaceae/química , Agrimonia/química , Antibacterianos/química , Línea Celular , Cólera/tratamiento farmacológico , Cólera/metabolismo , Toxina del Cólera/metabolismo , Fragaria/química , Gangliósido G(M1)/metabolismo , Humanos , Extractos Vegetales/química , Rubus/química , Vibrio cholerae/efectos de los fármacos , Vibrio cholerae/metabolismo
8.
Int J Mol Sci ; 20(12)2019 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-31212828

RESUMEN

Osteoblasts initiate bone mineralization by releasing matrix vesicles (MVs) into the extracellular matrix (ECM). MVs promote the nucleation process of apatite formation from Ca2+ and Pi in their lumen and bud from the microvilli of osteoblasts during bone development. Tissue non-specific alkaline phosphatase (TNAP) as well as annexins (among them, AnxA6) are abundant proteins in MVs that are engaged in mineralization. In addition, sarcoma proto-oncogene tyrosine-protein (Src) kinase and Rho-associated coiled-coil (ROCK) kinases, which are involved in vesicular transport, may also regulate the mineralization process. Upon stimulation in osteogenic medium containing 50 µg/mL of ascorbic acid (AA) and 7.5 mM of ß-glycerophosphate (ß-GP), human osteosarcoma Saos-2 cells initiated mineralization, as evidenced by Alizarin Red-S (AR-S) staining, TNAP activity, and the partial translocation of AnxA6 from cytoplasm to the plasma membrane. The addition of 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo [3,4-d] pyrimidine (PP2), which is an inhibitor of Src kinase, significantly inhibited the mineralization process when evaluated by the above criteria. In contrast, the addition of (R)-(+)-trans-4-(1-aminoethyl)-N-(4-pyridyl) cyclohexane carboxamide hydrochloride (Y-27632), which is an inhibitor of ROCK kinase, did not affect significantly the mineralization induced in stimulated Saos-2 cells as denoted by AR-S and TNAP activity. In conclusion, mineralization by human osteosarcoma Saos-2 cells seems to be differently regulated by Src and ROCK kinases.


Asunto(s)
Neoplasias Óseas/metabolismo , Calcificación Fisiológica , Osteosarcoma/metabolismo , Quinasas Asociadas a rho/metabolismo , Familia-src Quinasas/metabolismo , Anexinas/metabolismo , Biomarcadores , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Matriz Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Proto-Oncogenes Mas
9.
Biochim Biophys Acta Gen Subj ; 1862(3): 532-546, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29108957

RESUMEN

BACKGROUND: Matrix vesicles (MVs) are released from hypertrophic chondrocytes and from mature osteoblasts, the cells responsible for endochondral and membranous ossification. Under pathological conditions, they can also be released from cells of non-skeletal tissues such as vascular smooth muscle cells. MVs are extracellular vesicles of approximately 100-300nm diameter harboring the biochemical machinery needed to induce mineralization. SCOPE OF THE REVIEW: The review comprehensively delineates our current knowledge of MV biology and highlights open questions aiming to stimulate further research. The review is constructed as a series of questions addressing issues of MVs ranging from their biogenesis and functions, to biomimetic models. It critically evaluates experimental data including their isolation and characterization methods, like lipidomics, proteomics, transmission electron microscopy, atomic force microscopy and proteoliposome models mimicking MVs. MAJOR CONCLUSIONS: MVs have a relatively well-defined function as initiators of mineralization. They bind to collagen and their composition reflects the composition of lipid rafts. We call attention to the as yet unclear mechanisms leading to the biogenesis of MVs, and how minerals form and when they are formed. We discuss the prospects of employing upcoming experimental models to deepen our understanding of MV-mediated mineralization and mineralization disorders such as the use of reconstituted lipid vesicles, proteoliposomes and, native sample preparations and high-resolution technologies. GENERAL SIGNIFICANCE: MVs have been extensively investigated owing to their roles in skeletal and ectopic mineralization. MVs serve as a model system for lipid raft structures, and for the mechanisms of genesis and release of extracellular vesicles.


Asunto(s)
Condrocitos/ultraestructura , Matriz Extracelular/metabolismo , Vesículas Extracelulares , Osteoblastos/ultraestructura , Animales , Apatitas/metabolismo , Materiales Biomiméticos , Calcificación Fisiológica/fisiología , Calcinosis/fisiopatología , Condrocitos/patología , Colágeno/metabolismo , Vesículas Extracelulares/fisiología , Humanos , Hipertrofia , Microdominios de Membrana/fisiología , Minerales/metabolismo , Modelos Biológicos , Biogénesis de Organelos , Proteolípidos , Manejo de Especímenes , Calcificación Vascular/fisiopatología
10.
Postepy Biochem ; 64(3): 253-260, 2018 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-30656910

RESUMEN

Differentiation of cells of the skeletal tissue, such as osteoblasts and chondrocytes, into mineralization-competent cells is a necessary step of the physiological process of bone and cartilage mineralization. Vascular cell calcification accompanies a pathological process of atherosclerotic plaque formation, which occurs due to trans-differentiation of vascular smooth muscle cells into cells resembling bone mineralization-competent cells. The activity of tissue-nonspecific alkaline phosphatase (TNAP), an enzyme necessary for physiological mineralization, is also induced in vascular cells in response to inflammation. TNAP acquires its mineralizing function when anchored to the plasma membrane (PM) of mineralizing cells and to the surface of vesicles derived from these cells. Numerous important reports indicate that various types of vesicles play a crucial role in initiating cell differentiation. In this review, we would like to highlight various functions of different types of vesicular structures of the cellular transport machinery such as intracellular vesicles (IVs), extracellular vesicles (EVs) or matrix vesicles (MVs) at distinct stages of both physiological and pathological processes of tissue differentiation.


Asunto(s)
Huesos/citología , Cartílago/citología , Diferenciación Celular , Vesículas Citoplasmáticas/metabolismo , Vesículas Extracelulares/metabolismo , Calcificación Fisiológica , Matriz Extracelular , Humanos
11.
Biochim Biophys Acta Mol Basis Dis ; 1863(3): 643-653, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27932058

RESUMEN

Atherosclerotic plaque calcification varies from early, diffuse microcalcifications to a bone-like tissue formed by endochondral ossification. Recently, a paradigm has emerged suggesting that if the bone metaplasia stabilizes the plaques, microcalcifications are harmful. Tissue-nonspecific alkaline phosphatase (TNAP), an ectoenzyme necessary for mineralization by its ability to hydrolyze inorganic pyrophosphate (PPi), is stimulated by inflammation in vascular smooth muscle cells (VSMCs). Our objective was to determine the role of TNAP in trans-differentiation of VSMCs and calcification. In rodent MOVAS and A7R5 VSMCs, addition of exogenous alkaline phosphatase (AP) or TNAP overexpression was sufficient to stimulate the expression of several chondrocyte markers and induce mineralization. Addition of exogenous AP to human mesenchymal stem cells cultured in pellets also stimulated chondrogenesis. Moreover, TNAP inhibition with levamisole in mouse primary chondrocytes dropped mineralization as well as the expression of chondrocyte markers. VSMCs trans-differentiated into chondrocyte-like cells, as well as primary chondrocytes, used TNAP to hydrolyze PPi, and PPi provoked the same effects as TNAP inhibition in primary chondrocytes. Interestingly, apatite crystals, associated or not to collagen, mimicked the effects of TNAP on VSMC trans-differentiation. AP and apatite crystals increased the expression of BMP-2 in VSMCs, and TNAP inhibition reduced BMP-2 levels in chondrocytes. Finally, the BMP-2 inhibitor noggin blocked the rise in aggrecan induced by AP in VSMCs, suggesting that TNAP induction in VSMCs triggers calcification, which stimulates chondrogenesis through BMP-2. Endochondral ossification in atherosclerotic plaques may therefore be induced by crystals, probably to confer stability to plaques with microcalcifications.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Transdiferenciación Celular , Condrocitos/citología , Músculo Liso Vascular/citología , Animales , Calcificación Fisiológica , Calcio/metabolismo , Línea Celular , Condrocitos/metabolismo , Condrogénesis , Ratones , Músculo Liso Vascular/metabolismo , Placa Aterosclerótica/metabolismo
12.
Biochim Biophys Acta Gen Subj ; 1861(5 Pt A): 1009-1023, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28188861

RESUMEN

BACKGROUND: Members of Rho-associated coiled-coil kinases (ROCKs) are effectors of Rho family of small GTPases. ROCKs have multiple functions that include regulation of cellular contraction and polarity, adhesion, motility, proliferation, apoptosis, differentiation, maturation and remodeling of the extracellular matrix (ECM). SCOPE OF THE REVIEW: Here, we focus on the action of RhoA and RhoA effectors, ROCK1 and ROCK2, in cells related to tissue mineralization: mesenchymal stem cells, chondrocytes, preosteoblasts, osteoblasts, osteocytes, lining cells and osteoclasts. MAJOR CONCLUSIONS: The activation of the RhoA/ROCK pathway promotes stress fiber formation and reduces chondrocyte and osteogenic differentiations, in contrast to that in mesenchymal stem cells which stimulated the osteogenic and the chondrogenic differentiation. The effects of Rac1 and Cdc42 in promoting chondrocyte hypertrophy and of Rac1, Rac2 and Cdc42 in osteoclast are discussed. In addition, members of the Rho family of GTPases such Rac1, Rac2, Rac3 and Cdc42, acting upstream of ROCK and/or other protein effectors, may compensate the actions of RhoA, affecting directly or indirectly the actions of ROCKs as well as other protein effectors. GENERAL SIGNIFICANCE: ROCK activity can trigger cartilage degradation and affect bone formation, therefore these kinases may represent a possible therapeutic target to treat osteoarthritis and osseous diseases. Inhibition of Rho/ROCK activity in chondrocytes prevents cartilage degradation, stimulate mineralization of osteoblasts and facilitate bone formation around implanted metals. Treatment with osteoprotegerin results in a significant decrease in the expression of Rho GTPases, ROCK1 and ROCK2, reducing bone resorption. Inhibition of ROCK signaling increases osteoblast differentiation in a topography-dependent manner.


Asunto(s)
Calcificación Fisiológica/fisiología , Diferenciación Celular/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Osteoblastos/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Humanos
13.
Arch Biochem Biophys ; 593: 50-9, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26869201

RESUMEN

Mutations in the NPC1 or NPC2 genes lead to Niemann-Pick type C (NPC) disease, a rare lysosomal storage disorder characterized by progressive neurodegeneration. These mutations result in cholesterol and glycosphingolipid accumulation in the late endosomal/lysosomal compartment. Complications in the storage of cholesterol in NPC1 mutant cells are associated with other anomalies, such as altered distribution of intracellular organelles and properties of the plasma membrane. The pathomechanism of NPC disease is largely unknown. Interestingly, other storage diseases such as Gaucher and Farber diseases are accompanied by severe mitochondrial dysfunction. This prompted us to investigate the effect of absence or dysfunction of the NPC1 protein on mitochondrial properties to confirm or deny a putative relationship between NPC1 mutations and mitochondrial function. This study was performed on primary skin fibroblasts derived from skin biopsies of two NPC patients, carrying mutations in the NPC1 gene. We observed altered organization of mitochondria in NPC1 mutant cells, significant enrichment in mitochondrial cholesterol content, increased respiration, altered composition of the respiratory chain complex, and substantial reduction in cellular ATP level. Thus, a primary lysosomal defect in NPC1 mutant fibroblasts is accompanied by deregulation of the organization and function of the mitochondrial network.


Asunto(s)
Fibroblastos/metabolismo , Mitocondrias/fisiología , Enfermedades de Niemann-Pick/patología , Adenosina Trifosfato/biosíntesis , Adulto , Proteínas Portadoras/genética , Estudios de Casos y Controles , Colesterol/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Metabolismo Energético , Fibroblastos/ultraestructura , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Glicoproteínas de Membrana/genética , Potencial de la Membrana Mitocondrial , Mitocondrias/ultraestructura , Mutación , Proteína Niemann-Pick C1 , Fosforilación Oxidativa , Consumo de Oxígeno , Especies Reactivas de Oxígeno/metabolismo , Piel/patología
14.
Langmuir ; 32(48): 12923-12933, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27934520

RESUMEN

Nucleoside diphosphate kinases (NDPKs) are crucial elements in a wide array of cellular physiological or pathophysiological processes such as apoptosis, proliferation, or metastasis formation. Among the NDPK isoenzymes, NDPK-B, a cytoplasmic protein, was reported to be associated with several biological membranes such as plasma or endoplasmic reticulum membranes. Using several membrane models (liposomes, lipid monolayers, and supported lipid bilayers) associated with biophysical approaches, we show that lipid membrane binding occurs in a two-step process: first, initiation by a strong electrostatic adsorption process and followed by shallow penetration of the protein within the membrane. The NDPK-B binding leads to a decrease in membrane fluidity and formation of protein patches. The ability of NDPK-B to form microdomains at the membrane level may be related to protein-protein interactions triggered by its association with anionic phospholipids. Such accumulation of NDPK-B would amplify its effects in functional platform formation and protein recruitment at the membrane.


Asunto(s)
Membrana Celular/química , Membrana Dobles de Lípidos/química , Fluidez de la Membrana , Humanos , Nucleósido-Difosfato Quinasa/química , Unión Proteica
15.
Postepy Biochem ; 62(2): 216-223, 2016.
Artículo en Polaco | MEDLINE | ID: mdl-28132474

RESUMEN

Annexins form a family of membrane- and calcium-binding proteins, widely distributed in vertebrates. Their interactions with membranes are regulated by changes of intracellular concentration of calcium ([Ca2+]in.), pH, and the presence of negatively charged phospholipids as well as cholesterol in membranes. As protein participating in membrane fusion and sensors of a [Ca2+]in. Annexins may regulate various signaling pathways including patways involving protein kinase C (PKC isoforms. They also particpate in membrane repair mechanisms (along with actin cytoskeleton and S100 protein), in the vesicular transport (cholesterol enriched domains) as well in in intracellular calcium homeostasis and regulation of mitochondrial function and mitochondrial network structure. The last possibility is a topic of present review commemorating 90th Birthday of Professor Lech Wojtczak.


Asunto(s)
Anexinas/metabolismo , Mitocondrias/metabolismo , Animales , Anexinas/fisiología , Eucariontes/metabolismo , Humanos , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/fisiología
16.
Postepy Biochem ; 62(4): 511-517, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28132453

RESUMEN

Vascular calcification accompanies the pathological process of atherosclerotic plaque formation. Artery calcification results from trans-differentiation of vascular smooth muscle cells (VSMCs) into cells resembling mineralization-competent cells such as osteoblasts and chondrocytes. The activity of tissue-nonspecific alkaline phosphatase (TNAP), a GPI-anchored enzyme necessary for physiological mineralization, is induced in VSMCs in response to inflammation. TNAP achieves its mineralizing function being anchored to plasma membrane of mineralizing cells and to the surface of their derived matrix vesicles (MVs), and numerous important reports indicate that membranes play a crucial role in initiating the crystal formation. In this review, we would like to highlight various functions of lipids and proteins associated to membranes at different stages of both physiological mineralization and vascular calcification, with an emphasis on the pathological process of atherosclerotic plaque formation.


Asunto(s)
Calcinosis , Placa Aterosclerótica/metabolismo , Animales , Calcificación Fisiológica , Condrocitos , Humanos , Lípidos de la Membrana , Osteoblastos , Placa Aterosclerótica/fisiopatología
17.
Postepy Biochem ; 61(4): 430-5, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27048097

RESUMEN

One of the main factors causing bacterial diarrhea are AB5 enterotoxins. This group is divided into four families: pertussis toxin, cholera toxin, shiga toxin and subtilase cytotoxin. In this review we will describe the activity, structure and function of the cholera and shiga toxin families. The AB5 enterotoxins contain a catalytic subunit A and pentameric subunit B, which binds to the cell surface within lipid rafts. The cholera toxin family cause the constitutive activation of Gsa protein, which results in cAMP production, an opening of the chloride channels and releases chloride ions into the lumen of the small intestine. In contrast, the shiga toxin family has a cytotoxic effect on epithelial cells. It can inhibit protein synthesis leading to cell death. Although AB5 has a toxic activity, the B5 subunits have a significant potential as a transporter for proteins with anticancer activity and as a tool for the visualization of lipid rafts and cancer cells.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Enterotoxinas/química , Enterotoxinas/metabolismo , Infecciones Bacterianas/microbiología , Diarrea/microbiología , Humanos , Conformación Proteica
18.
Biogerontology ; 15(1): 47-64, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24243065

RESUMEN

Senescence of vascular smooth muscle cells (VSMCs) contributes to aging as well as age-related diseases of the cardiovascular system. Senescent VSMCs have been shown to be present in atherosclerotic plaques. Both replicative (RS) and stress-induced premature senescence (SIPS) accompany cardiovascular diseases. We aimed to establish the signature of RS and SIPS of VSMCs, induced by a common anticancer drug, doxorubicin, and to discover the so far undisclosed features of senescent cells that are potentially harmful to the organism. Most of the senescence hallmarks were common for both RS and SIPS; however, some differences were observed. 32 % of doxorubicin-treated cells were arrested in the G2/M phase of the cell cycle, while 73 % of replicatively senescing cells were arrested in the G1 phase. Moreover, on the basis of alkaline phosphatase activity measurements, we show that a 7-day treatment with doxorubicin (dox), does not cause precocious cell calcification, which is a characteristic feature of RS. We did not observe calcification even though after 7 days of dox-treatment many other markers characteristic for senescent cells were present. It can suggest that dox-induced SIPS does not accelerate the mineralization of vessels. We consider that detailed characterization of the two types of cellular senescence can be useful in in vitro studies of potential anti-aging factors.


Asunto(s)
Envejecimiento Prematuro/inducido químicamente , Envejecimiento Prematuro/patología , Aorta/citología , Proliferación Celular , Senescencia Celular/fisiología , Doxorrubicina/efectos adversos , Músculo Liso Vascular/citología , Envejecimiento Prematuro/fisiopatología , Fosfatasa Alcalina/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Línea Celular , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Humanos , Técnicas In Vitro , Masculino , Músculo Liso Vascular/fisiología , Superóxidos/metabolismo , Homeostasis del Telómero/fisiología , Adulto Joven , beta-Galactosidasa/metabolismo
19.
Postepy Biochem ; 60(1): 62-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25033543

RESUMEN

Cancer cells degrade the extracellular matrix (ECM) in the basement membrane and blood vessel walls to emigrate and invade from original to peripheral tissues. This invasion of cells through ECM layers is a key step not only in tumor metastasis but also in other processes such as inflammation and development. All of them seem to be facilitated by the formation of small cellular protrusions of localized protease activity, termed podosomes in non-malignant cells and invadopodia in cancer cells. Understanding the mechanisms that lead to functional invadopodia is nowadays a subject of intense study. Herein, a brief overview of the molecular components and regulators of invadopodia will be provided. In this review we will summarize recent achievements and the latest methods of visualizing invadopodia formation and functions, with a strong emphasis on advanced microscopy approaches.


Asunto(s)
Extensiones de la Superficie Celular/metabolismo , Matriz Extracelular/metabolismo , Invasividad Neoplásica/fisiopatología , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Membrana Basal/metabolismo , Movimiento Celular , Extensiones de la Superficie Celular/patología , Exocitosis/fisiología , Humanos , Transducción de Señal , Proteínas de Transporte Vesicular/metabolismo
20.
Org Biomol Chem ; 11(32): 5332-8, 2013 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-23842795

RESUMEN

Fluorescent analogues provide important tools for biochemical/biophysical research. However, the analogues contain chemical modifications much larger than those known to affect ligand-binding, such as the inversion of a carbon centre or substitution of an atom. We lack experimental tools and protocols to select the most appropriate fluorescent analogue. Herein, we use several NMR spectroscopy methods, including Saturation Transfer Difference (STD), STD competition and transferred nuclear Overhauser effect spectroscopy (Tr-NOESY), as tools to select appropriate fluorescent probes. Annexin A6 (AnxA6) is a ubiquitous protein that forms in vitro GTP-induced ion channels. We used this protein as a model and screened guanosine triphosphate (GTP) and four fluorescent analogues against AnxA6. STD reported that the GTP moiety of all ligands made similar contacts with the protein, despite additional interactions between the fluorescent tags and AnxA6. Competition STD experiments verified that the analogues and GTP bind to the same site. Tr-NOESY indicated that the bound conformation of the base relative to ribose is altered for some analogues compared to GTP. MANT-GTP or the BODIPY thioester of guanosine 5'-O-(3-thiotriphosphate) are the most suitable fluorescent analogues for AnxA6, according to NMR. These results reveal NMR as a useful technique to select and design proper fluorescent tags for biochemical/biophysical assays.


Asunto(s)
Colorantes Fluorescentes/química , Espectroscopía de Resonancia Magnética/métodos , Nucleótidos/química , Anexina A6/análisis , Anexina A6/metabolismo , Colorantes Fluorescentes/metabolismo , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/metabolismo , Humanos , Nucleótidos/metabolismo , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA