Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Langmuir ; 36(19): 5134-5144, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32338922

RESUMEN

Lipid-based carriers such as liposomes represent one of the most advanced classes of drug delivery systems. Their clinical success relies on their composition, similar to that of the cell membrane. Their cellular specificity often relies on a ligand-receptor interaction. Although differences in the physicochemical properties of the cell membrane between tumor and nontumor cells have been reported, they are not systematically used for drug delivery purposes. In this report, a new approach was developed to ensure selective targeting based on physical compatibility between the target and the carrier membranes. By modulating the liposome composition and thus its membrane fluidity, we achieved selective targeting on four cancer cell lines of varying aggressiveness. Furthermore, using membrane-embedded and inner core-encapsulated fluorophores, we assessed the mechanism of this interaction to be based on the fusion of the liposome with the cell membranes. Membrane fluidity is therefore a major parameter to be considered when designing lipid drug carriers as a promising, lower cost alternative to current targeting strategies based on covalent grafting.


Asunto(s)
Fluidez de la Membrana , Neoplasias , Sistemas de Liberación de Medicamentos , Humanos , Lípidos , Liposomas , Neoplasias/tratamiento farmacológico
2.
Mol Cell Biochem ; 473(1-2): 263-279, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32661773

RESUMEN

Prostate cancer (PCa) is the most frequent cancer among men and the first cause of death over 65. Approximately 90% of patients with advanced disease will develop bone metastasis, which dramatically reduces long-term survival. Therefore, effective therapies need to be developed, especially when disease is still well-localized. Phospholipase D (PLD), an enzyme that hydrolyzes phosphatidylcholine to yield phosphatidic acid, regulates several cellular functions as proliferation, survival, migration or vesicular trafficking. PLD is implicated in numerous diseases such as neurodegenerative, cardiovascular, autoimmune disorders or cancer. Indeed, PLD controls different aspects of oncogenesis including tumor progression and resistance to targeted therapies such as radiotherapy. PLD1 and PLD2 are the only isoforms with catalytic activity involved in cancer. Surprisingly, studies deciphering the role of PLD in the pathophysiology of PCa are scarce. Here we describe the correlation between PLD activity and PLD1 and PLD2 expression in PCa bone metastasis-derived cell lines C4-2B and PC-3. Next, by using PLD pharmacological inhibitors and RNA interference strategy, we validate the implication of PLD1 and PLD2 in cell viability, clonogenicity and proliferation of C4-2B and PC-3 cells and in migration capacity of PC-3 cells. Last, we show an increase in PLD activity as well as PLD2 protein expression during controlled starvation of PC-3 cells, concomitant with an augmentation of its migration capacity. Specifically, upregulation of PLD activity appears to be PKC-independent. Taken together, our results indicate that PLD, and in particular PLD2, could be considered as a potential therapeutic target for the treatment of PCa-derived bone metastasis.


Asunto(s)
Carcinogénesis/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfolipasa D/metabolismo , Neoplasias de la Próstata/enzimología , Carcinogénesis/genética , Carcinogénesis/patología , Humanos , Masculino , Proteínas de Neoplasias/genética , Células PC-3 , Fosfolipasa D/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología
3.
J Cell Physiol ; 234(4): 4825-4839, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30207376

RESUMEN

Vascular calcification (VC) is the pathological accumulation of calcium phosphate crystals in one of the layers of blood vessels, leading to loss of elasticity and causing severe calcification in vessels. Medial calcification is mostly seen in patients with chronic kidney disease (CKD) and diabetes. Identification of key enzymes and their actions during calcification will contribute to understand the onset of pathological calcification. Phospholipase D (PLD1, PLD2) is active at the earlier steps of mineralization in osteoblasts and chondrocytes. In this study, we aimed to determine their effects during high-phosphate treatment in mouse vascular smooth muscle cell line MOVAS, in the ex vivo model of the rat aorta, and in the in vivo model of adenine-induced CKD. We observed an early increase in PLD1 gene and protein expression along with the increase in the PLD activity in vascular muscle cell line, during calcification induced by ascorbic acid and ß-glycerophosphate. Inhibition of PLD1 by the selective inhibitor VU0155069, or the pan-PLD inhibitor, halopemide, prevented calcification. The mechanism of PLD activation is likely to be protein kinase C (PKC)-independent since bisindolylmaleimide X hydrochloride, a pan-PKC inhibitor, did not affect the PLD activity. In agreement, we found an increase in Pld1 gene expression and PLD activity in aortic explant cultures treated with high phosphate, whereas PLD inhibition by halopemide decreased calcification. Finally, an increase in both Pld1 and Pld2 expression occurred simultaneously with the appearance of VC in a rat model of CKD. Thus, PLD, especially PLD1, promotes VC in the context of CKD and could be an important target for preventing onset or progression of VC.


Asunto(s)
Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Fosfolipasa D/metabolismo , Fósforo Dietético , Insuficiencia Renal Crónica/complicaciones , Calcificación Vascular/etiología , Animales , Calcio de la Dieta , Línea Celular , Transdiferenciación Celular , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Inhibidores de Fosfodiesterasa/farmacología , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/genética , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/enzimología , Transducción de Señal , Técnicas de Cultivo de Tejidos , Calcificación Vascular/enzimología , Calcificación Vascular/patología , Calcificación Vascular/prevención & control
4.
J Cell Biochem ; 120(4): 5923-5935, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30320913

RESUMEN

Mammalian phospholipase D (PLD) mostly hydrolyzes phosphatidylcholine producing phosphatidic acid. PLD activity was previously detected in different osteoblastic cell models, and was increased by several growth factors involved in bone homeostasis. To confirm possible actions of PLD isoforms during mineralization process, we analyzed their effects in osteoblastic cell models and during bone formation. PLD1 expression, along with PLD activity, increased during differentiation of primary osteoblasts and Saos-2 cells, and peaked at the onset of mineralization. Subsequently, both PLD1 expression and PLD activity decreased, suggesting that PLD1 function is regulated during osteoblast maturation. In contrast, PLD2 expression was not significantly affected during differentiation of osteoblasts. Overexpression of PLD1 in Saos-2 cells improved their mineralization potential. PLD inhibitor Halopemide or PLD1-selective inhibitor, led to a decrease in mineralization in both cell types. On the contrary, the selective inhibitor of PLD2, did not affect the mineralization process. Moreover, primary osteoblasts isolated from PLD1 knockout (KO) mice were significantly less efficient in mineralization as compared with those isolated from wild type (WT) or PLD2 KO mice. In contrast, bone formation, as monitored by high-resolution microcomputed tomography analysis, was not impaired in PLD1 KO nor in PLD2 KO mice, indicating that the lack of PLD1 or that of PLD2 did not affect the bone structure in adult mice. Taken together, our findings indicate that PLD activity, especially which of PLD1 isoform, may enhance the mineralization process in osteoblastic cells. Nonetheless, the lack of PLD1 or PLD2 do not seem to significantly affect bone formation in adult mice.


Asunto(s)
Osteoblastos/metabolismo , Fosfolipasa D/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Western Blotting , Calcificación Fisiológica/fisiología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Femenino , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteogénesis/fisiología , Fosfolipasa D/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Bioorg Med Chem ; 27(6): 1034-1042, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30773420

RESUMEN

Selective proteinase inhibitors have demonstrated utility in the investigation of cartilage degeneration mechanisms and may have clinical use in the management of osteoarthritis. The cysteine protease cathepsin K (CatK) is an attractive target for arthritis therapy. Here we report the synthesis of two cathepsin K inhibitors (CKIs): racemic azanitrile derivatives CKI-E and CKI-F, which have better inhibition properties on CatK than the commercial inhibitor odanacatib (ODN). Their IC50 values and inhibition constants (Ki) have been determined in vitro. Inhibitors demonstrate differential selectivity for CatK over cathepsin B, L and S in vitro, with Ki amounting to 1.14 and 7.21 nM respectively. We analyzed the effect of these racemic inhibitors on viability in different cell types. The human osteoblast-like cell line MG63, MOVAS cells (a murine vascular smooth muscle cell line) or murine primary chondrocytes, were treated either with CKI-E or with CKI-F, which were not toxic at doses of up to 5 µM. Primary chondrocytes subjected to several passages were used as a model of phenotypic loss of articular chondrocytes, occurring in osteoarthritic cartilage. The efficiency of CKIs regarding CatK inhibition and their specificity over other proteases were validated in primary chondrocytes subjected to several passages. Racemic CKI-E and CKI-F at 0.1 and 1 µM significantly inhibited CatK activity in dedifferentiated chondrocytes, even better than the commercial CatK inhibitor ODN. The enzymatic activity of other proteases such as matrix metalloproteinases or aggrecanases were not affected. Taken together, these findings support the possibility to design CatK inhibitors for preventing cartilage degradation in different pathologies.


Asunto(s)
Catepsina K/antagonistas & inhibidores , Desdiferenciación Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Nitrilos/farmacología , Inhibidores de Proteasas/farmacología , Animales , Compuestos Aza/síntesis química , Compuestos Aza/química , Compuestos Aza/farmacología , Catepsina K/metabolismo , Línea Celular , Células Cultivadas , Condrocitos/citología , Condrocitos/enzimología , Diseño de Fármacos , Humanos , Ratones , Nitrilos/síntesis química , Nitrilos/química , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química
6.
Bioorg Med Chem ; 22(17): 4955-60, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25047939

RESUMEN

The bioactive metabolite sphingosine-1-phosphate (S1P), a product of sphingosine kinases (SphKs), mediates diverse biological processes such as cell differentiation, proliferation, survival and angiogenesis. A fluorinated analogue of S1P receptor agonist has been synthesized by utilizing a ring opening reaction of oxacycles by a lithiated difluoromethylphosphonate anion as the key reaction. In vitro activity of this S1P analogue is also reported.


Asunto(s)
Organofosfatos/síntesis química , Organofosfatos/farmacología , Receptores de Lisoesfingolípidos/agonistas , Esfingosina/análogos & derivados , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Halogenación , Humanos , Masculino , Estructura Molecular , Organofosfatos/química , Esfingosina/síntesis química , Esfingosina/química , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato , Relación Estructura-Actividad
7.
Cancers (Basel) ; 15(10)2023 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-37345069

RESUMEN

Prostate cancer (PC) is the second most common cancer in men worldwide. More than 65% of men diagnosed with PC are above 65. Patients with localized PC show high long-term survival, however with the disease progression into a metastatic form, it becomes incurable, even after strong radio- and/or chemotherapy. Sphingosine 1-phosphate (S1P) is a bioactive lipid that participates in all the steps of oncogenesis including tumor cell proliferation, survival, migration, invasion, and metastatic spread. The S1P-producing enzymes sphingosine kinases 1 and 2 (SK1 and SK2), and the S1P degrading enzyme S1P lyase (SPL), have been shown to be highly implicated in the onset, development, and therapy resistance of PC during the last 20 years. In this review, the most important studies demonstrating the role of S1P and S1P metabolic partners in PC are discussed. The different in vitro, ex vivo, and in vivo models of PC that were used to demonstrate the implication of S1P metabolism are especially highlighted. Furthermore, the most efficient molecules targeting S1P metabolism that are under preclinical and clinical development for curing PC are summarized. Finally, the possibility of targeting S1P metabolism alone or combined with other therapies in the foreseeable future as an alternative option for PC patients is discussed. Research Strategy: PubMed from INSB was used for article research. First, key words "prostate & sphingosine" were used and 144 articles were found. We also realized other combinations of key words as "prostate cancer bone metastasis" and "prostate cancer treatment". We used the most recent reviews to illustrate prostate cancer topic and sphingolipid metabolism overview topic.

8.
Biomolecules ; 14(1)2023 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-38254642

RESUMEN

Mineralization-competent cells, including hypertrophic chondrocytes, mature osteoblasts, and osteogenic-differentiated smooth muscle cells secrete media extracellular vesicles (media vesicles) and extracellular vesicles bound to the extracellular matrix (matrix vesicles). Media vesicles are purified directly from the extracellular medium. On the other hand, matrix vesicles are purified after discarding the extracellular medium and subjecting the cells embedded in the extracellular matrix or bone or cartilage tissues to an enzymatic treatment. Several pieces of experimental evidence indicated that matrix vesicles and media vesicles isolated from the same types of mineralizing cells have distinct lipid and protein composition as well as functions. These findings support the view that matrix vesicles and media vesicles released by mineralizing cells have different functions in mineralized tissues due to their location, which is anchored to the extracellular matrix versus free-floating.


Asunto(s)
Calcinosis , Vesículas Extracelulares , Humanos , Matriz Extracelular , Condrocitos , Hipertrofia
9.
J Biomed Mater Res B Appl Biomater ; 110(2): 338-349, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34289232

RESUMEN

A new generation of ceramic on ceramic (BIOLOX ®delta) bearings has emerged more than 10 years ago proving a high resistance to wear and good clinical results. However, biological reactions to wear debris, particularly the nanoparticles, need to be evaluated. The first originality of this study is to start from real wear particles obtained by the hip walking simulator (CERsim). These particles were compared with particles obtained by usual methods to assess the biocompatibility of materials: press machine (CERpress). Two ranges of ceramic particles were thus observed: ceramic particles with micron (intergranular fractures) and nano sizes (intragranular fractures), and characterized compared to ultra-high molecular weight polyethylene (UHMWPE). The second originality of this work is to assess the cellular reaction using the primary joint chondrocyte cultures simulating the osteogenesis process and not the cell lines, which are used to simulate the biological reaction of osteolysis. The first results showed a significant difference in cell viability between the cells in contact with particles from the walking simulator and those obtained with the press machine. On the other hand, it was found that the way of extraction of the particles from the lubricant could significantly affect the biological reaction. More interestingly, nano-sized ceramic particles showed a significant impact on the secretion of functional inflammatory mediators, agreeing with recent results in vivo. These novel methods of characterizing the osteogenic impact of UHMWPE and ceramic wear debris can complement the conventional expertise method focusing previously on the osteolysis aspect.


Asunto(s)
Prótesis de Cadera , Cerámica , Condrocitos , Prótesis de Cadera/efectos adversos , Humanos , Ensayo de Materiales , Osteogénesis , Polietilenos , Falla de Prótesis , Caminata
10.
FASEB J ; 24(10): 3882-94, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20522783

RESUMEN

The sphingosine kinase-1/sphingosine 1-phosphate (SphK1/S1P) pathway has been associated with cancer promotion and progression and resistance to treatments in a number of cancers, including prostate adenocarcinoma. Here we provide the first evidence that dietary agents, namely, epigallocatechin gallate (EGCg, IC(50)≈75 µM), resveratrol (IC(50)≈40 µM), or a mixture of polyphenols from green tea [polyphenon E (PPE), IC(50)≈70 µM] or grapevine extract (vineatrol, IC(50)≈30 µM), impede prostate cancer cell growth in vitro and in vivo by inhibiting the SphK1/S1P pathway. We establish that SphK1 is a downstream effector of the ERK/phospholipase D (PLD) pathway, which is inhibited by green tea and wine polyphenols. Enforced expression of SphK1 impaired the ability of green tea and wine polyphenols, as well as pharmacological inhibitors of PLD and ERK activities, to induce apoptosis in PC-3 and C4-2B cells. The therapeutic efficacy of these polyphenols on tumor growth and the SphK1/S1P pathway were confirmed in animals using a heterotopic PC-3 tumor in place model. PC-3/SphK1 cells implanted in animals developed larger tumors and resistance to treatment with polyphenols. Furthermore, using an orthotopic PC-3/GFP model, the chemopreventive effect of an EGCg or PPE diet was associated with SphK1 inhibition, a decrease in primary tumor volume, and occurrence and number of metastases. These results provide the first demonstration that the prosurvival, antiapoptotic SphK1/S1P pathway represents a target of dietary green tea and wine polyphenols in cancer.


Asunto(s)
Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Neoplasias de la Próstata/patología , Té/química , Vino/análisis , Humanos , Masculino
11.
Int J Biol Macromol ; 166: 1131-1140, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33161081

RESUMEN

Phospholipase D (PLD) is a ubiquitous enzyme that cleaves the distal phosphoester bond of phospholipids generating phosphatidic acid (PA). In plants, PA is involved in numerous cell responses triggered by stress. Similarly, in mammals, PA is also a second messenger involved in tumorigenesis. PLD is nowadays considered as a therapeutic target and blocking its activity with specific inhibitors constitutes a promising strategy to treat cancers. Starting from already described PLD inhibitors, this study aims to investigate the effect of their structural modifications on the enzyme's activity, as well as identifying new potent inhibitors of eukaryotic PLDs. Being able to purify the plant PLD from Vigna unguiculata (VuPLD), we obtained a SAXS model of its structure. We then used a fluorescence-based test suitable for high-throughput screening to review the effect of eukaryotic PLD inhibitors described in the literature. In this regard, we found that only few molecules were in fact able to inhibit VuPLD and we confirmed that vanadate is the most potent of all with an IC50 around 58 µM. Moreover, the small-scale screening of a chemical library of 3120 compounds allowed us to optimize the different screening's steps and paved the way towards the discovery of new potent inhibitors.


Asunto(s)
Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/análisis , Inhibidores Enzimáticos/farmacología , Fosfolipasa D/antagonistas & inhibidores , Alcoholes/farmacología , Ensayos Analíticos de Alto Rendimiento , Humanos , Hidrólisis , Fosfolipasa D/metabolismo , Sales (Química)/farmacología , Dispersión del Ángulo Pequeño , Vanadatos/farmacología , Vigna/enzimología , Difracción de Rayos X
12.
Mol Cancer Ther ; 8(4): 809-20, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19372554

RESUMEN

Defeating pancreatic cancer resistance to the chemotherapeutic drug gemcitabine remains a challenge to treat this deadly cancer. Targeting the sphingolipid metabolism for improving tumor chemosensitivity has recently emerged as a promising strategy. The fine balance between intracellular levels of the prosurvival sphingosine-1-phosphate (S1P) and the proapoptotic ceramide sphingolipids determines cell fate. Among enzymes that control this metabolism, sphingosine kinase-1 (SphK1), a tumor-associated protein overexpressed in many cancers, favors survival through S1P production, and inhibitors of SphK1 are used in ongoing clinical trials to sensitize epithelial ovarian and prostate cancer cells to various chemotherapeutic drugs. We here report that the cellular ceramide/S1P ratio is a critical biosensor for predicting pancreatic cancer cell sensitivity to gemcitabine. A low level of the ceramide/S1P ratio, associated with a high SphK1 activity, correlates with a robust intrinsic pancreatic cancer cell chemoresistance toward gemcitabine. Strikingly, increasing the ceramide/S1P ratio, by using pharmacologic (SphK1 inhibitor or ceramide analogue) or small interfering RNA-based approaches to up-regulate intracellular ceramide levels or reduce SphK1 activity, sensitized pancreatic cancer cells to gemcitabine. Conversely, decreasing the ceramide/S1P ratio, by up-regulating SphK1 activity, promoted gemcitabine resistance in these cells. Development of novel pharmacologic strategies targeting the sphingolipid metabolism might therefore represent an interesting promising approach, when combined with gemcitabine, to defeat pancreatic cancer chemoresistance to this drug.


Asunto(s)
Ceramidas/metabolismo , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos , Lisofosfolípidos/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/análogos & derivados , Western Blotting , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Desoxicitidina/farmacología , Inhibidores Enzimáticos/farmacología , Humanos , Neoplasias Pancreáticas/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleótido Reductasas/antagonistas & inhibidores , Esfingosina/metabolismo , Células Tumorales Cultivadas , Gemcitabina
13.
Biochim Biophys Acta Mol Basis Dis ; 1866(12): 165919, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32800947

RESUMEN

Prostate cancer (PCa) is the most frequent cancer in men aged 65 and over. PCa mainly metastasizes in the bone, forming osteosclerotic lesions, inducing pain, fractures, and nerve compression. Cancer cell-derived exosomes participate in the metastatic spread, ranging from oncogenic reprogramming to the formation of pre-metastatic niches. Moreover, exosomes were recently involved in the dialog between PCa cells and the bone metastasis microenvironment. Phospholipase D (PLD) isoforms PLD1/2 catalyze the hydrolysis of phosphatidylcholine to yield phosphatidic acid (PA), regulating tumor progression and metastasis. PLD is suspected to play a role in exosomes biogenesis. We aimed to determine whether PCa-derived exosomes, through PLD, interact with the bone microenvironment, especially osteoblasts, during the metastatic process. Here we demonstrate for the first time that PLD2 is present in exosomes of C4-2B and PC-3 cells. C4-2B-derived exosomes activate proliferation and differentiation of osteoblasts models, by stimulating ERK 1/2 phosphorylation, by increasing the tissue-nonspecific alkaline phosphatase activity and the expression of osteogenic differentiation markers. Contrariwise, when C4-2B exosomes are generated in the presence of halopemide, a PLD pan-inhibitor, they lose their ability to stimulate osteoblasts. Furthermore, the number of released exosomes diminishes significantly (-40%). When the PLD product PA is combined with halopemide, exosome secretion is fully restored. Taken together, our results indicate that PLD2 stimulates exosome secretion in PCa cell models as well as their ability to increase osteoblast activity. Thus, PLD2 could be considered as a potent player in the establishment of PCa bone metastasis acting through tumor cell derived-exosomes.


Asunto(s)
Diferenciación Celular , Osteoblastos/citología , Osteoblastos/metabolismo , Fosfolipasa D/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Células 3T3 , Animales , Células Cultivadas , Exosomas/metabolismo , Humanos , Masculino , Ratones
14.
Bone ; 130: 115087, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31648078

RESUMEN

Sphingolipids display important functions in various pathologies such as cancer, obesity, diabetes, cardiovascular or neurodegenerative diseases. Sphingosine, sphingosine 1-phosphate (S1P), and ceramide are the central molecules of sphingolipid metabolism. Sphingosine kinases 1 and 2 (SK1 and SK2) catalyze the conversion of the sphingolipid metabolite sphingosine into S1P. The balance between the levels of S1P and its metabolic precursors ceramide and sphingosine has been considered as a switch that could determine whether a cell proliferates or dies. This balance, also called « sphingolipid rheostat ¼, is mainly under the control of SKs. Several studies have recently pointed out the contribution of SK/S1P metabolic pathway in skeletal development, mineralization and bone homeostasis. Indeed, SK/S1P metabolism participates in different diseases including rheumatoid arthritis, spondyloarthritis, osteoarthritis, osteoporosis, cancer-derived bone metastasis or calcification disorders as vascular calcification. In this review, we will summarize the most important data regarding the implication of SK/S1P axis in bone and joint diseases and ectopic calcification, and discuss the therapeutic potential of targeting SK/S1P metabolism for the treatment of these pathologies.


Asunto(s)
Neoplasias , Espondiloartritis , Humanos , Lisofosfolípidos , Esfingosina/análogos & derivados
15.
J Bone Miner Res ; 34(12): 2264-2276, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31373726

RESUMEN

Spondyloarthritis (SpA) is a common rheumatic disease characterized by enthesis inflammation (enthesitis) and ectopic ossification (enthesophytes). The current pathogenesis model suggests that inflammation and mechanical stress are both strongly involved in SpA pathophysiology. We have previously observed that the levels of sphingosine 1-phosphate (S1P), a bone anabolic molecule, were particularly high in SpA patients' serum compared to healthy donors. Therefore, we wondered how this deregulation was related to SpA molecular mechanisms. Mouse primary osteoblasts, chondrocytes, and tenocytes were used as cell culture models. The sphingosine kinase 1 (Sphk1) gene expression and S1P secretion were significantly enhanced by cyclic stretch in osteoblasts and chondrocytes. Further, TNF-α and IL-17, cytokines implicated in enthesitis, increased Sphk1 mRNA in chondrocytes in an additive manner when combined to stretch. The immunochemistry on mouse ankles showed that sphingosine kinase 1 (SK1) was localized in some chondrocytes; the addition of a pro-inflammatory cocktail augmented Sphk1 expression in cultured ankles. Subsequently, fingolimod was used to block S1P metabolism in cell cultures. It inhibited S1P receptors (S1PRs) signaling and SK1 and SK2 activity in both osteoblasts and chondrocytes. Fingolimod also reduced S1PR-induced activation by SpA patients' synovial fluid (SF), demonstrating that the stimulation of chondrocytes by SFs from SpA patients involves S1P. In addition, when the osteogenic culture medium was supplemented with fingolimod, alkaline phosphatase activity, matrix mineralization, and bone formation markers were significantly reduced in osteoblasts and hypertrophic chondrocytes. Osteogenic differentiation was accompanied by an increase in S1prs mRNA, especially S1P1/3 , but their contribution to S1P-impact on mineralization seemed limited. Our results suggest that S1P might be overproduced in SpA enthesis in response to cytokines and mechanical stress, most likely by chondrocytes. Moreover, S1P could locally favor the abnormal ossification of the enthesis; therefore, blocking the S1P metabolic pathway could be a potential therapeutic approach for the treatment of SpA. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Citocinas/farmacología , Lisofosfolípidos/biosíntesis , Osteogénesis , Esfingosina/análogos & derivados , Espondiloartritis/patología , Espondiloartritis/fisiopatología , Estrés Mecánico , Adolescente , Adulto , Anciano , Animales , Calcificación Fisiológica/efectos de los fármacos , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Femenino , Clorhidrato de Fingolimod/farmacología , Humanos , Masculino , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Persona de Mediana Edad , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Esfingosina/biosíntesis , Líquido Sinovial/metabolismo , Tenocitos/efectos de los fármacos , Tenocitos/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Adulto Joven
16.
Bone ; 103: 150-158, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28684192

RESUMEN

Spondyloarthritis (SpA) is a relatively common chronic inflammatory joint disorder, with a prevalence of about 0.2-0.5% worldwide. The primary target of the pathological process is the enthesis, where tendons and ligaments attach to underlying bone. These insertion sites are hotspots of bone formation (enthesophytes), which can lead to ankylosis. Unfortunately, the mechanisms causing the onset and progression of entheseal ossification remain largely unknown. Sphingosine 1-phosphate (S1P), a lipid generated after sphingosine phosphorylation by sphingosine kinases 1 and 2 (SK1/2), plays important roles in cell proliferation, differentiation and survival. S1P regulates fundamental biological processes such as cell cycle, inflammatory response or bone homeostasis. Indeed, S1P has been involved in some of most-spread skeletal diseases such as rheumatoid arthritis or osteoarthritis. On the other hand, the implication of S1P in SpA has not been explored yet. In the present work, we observed by ELISA that S1P content was significantly increased in the serum of SpA patients (6.1±4.2µM, n=21) compared to healthy donors (1.6±0.9µM, n=12). In vitro, gene expression of SK1 and SK2 as well as their activity were increased during differentiation of primary murine chondrocytes and osteoblasts into mineralizing cells. In addition, mRNA of the S1P-specific transporter Spns2 and S1P secretion were augmented. Using the pharmacological drugs SKi (SK pan-inhibitor), PF-543 (SK1 specific inhibitor) or K-145 (SK2 specific inhibitor), we showed that the inhibition of SK1 and/or SK2 decreased matrix mineralization, alkaline phosphatase activity and the mRNA expression of Runx2 and Bglap in chondrocytes and osteoblasts. To our knowledge, this is the first study indicating that S1P levels are significantly increased in serum from SpA patients. Moreover, we showed in vitro that SK activity was involved in the mineralization capacity of osteoblasts and chondrocytes. S1P metabolic pathway may represent an ingenious therapeutic target for SpA in the future.


Asunto(s)
Lisofosfolípidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/análogos & derivados , Espondiloartritis/metabolismo , Adolescente , Adulto , Anciano , Animales , Calcificación Fisiológica/fisiología , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Esfingosina/metabolismo , Adulto Joven
17.
FEBS Lett ; 535(1-3): 101-5, 2003 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-12560086

RESUMEN

We show here for the first time that sphingosine-1-phosphate (Sph-1-P) stimulates cortisol secretion in zona fasciculata cells of bovine adrenal glands. This effect was dependent upon protein kinase C (PKC) and extracellular Ca2+, and was inhibited by pertussis toxin. Sph-1-P activated phospholipase D (PLD) through a pertussis toxin-sensitive mechanism, also involving extracellular Ca2+ and PKC. Primary alcohols, which attenuate formation of phosphatidic acid (the product of PLD), and cell-permeable ceramides, which inhibit PLD, blocked Sph-1-P-induced cortisol secretion. In conclusion, Sph-1-P stimulates cortisol secretion through a mechanism involving Gi/o protein-coupled receptors, extracellular Ca2+, PKC and PLD.


Asunto(s)
Hidrocortisona/metabolismo , Lisofosfolípidos , Esfingosina/análogos & derivados , Esfingosina/farmacología , Zona Fascicular/efectos de los fármacos , Zona Fascicular/metabolismo , Animales , Calcio/metabolismo , Bovinos , Células Cultivadas , Ceramidas/farmacología , Quelantes/farmacología , Activación Enzimática/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Lípidos/farmacología , Toxina del Pertussis/farmacología , Ácidos Fosfatidicos/biosíntesis , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Zona Fascicular/citología
18.
Mol Cell Endocrinol ; 222(1-2): 9-20, 2004 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-15249121

RESUMEN

Angiotensin II (Ang-II) regulates a variety of cellular functions including cortisol secretion. In the present report, we demonstrate that Ang-II activates phospholipase D (PLD) in zona fasciculata (ZF) cells of bovine adrenal glands, and that this effect is associated to the stimulation of cortisol secretion by this hormone. PLD activation was dependent upon extracellular Ca2+, and was blocked by inhibition of protein kinase C (PKC). Using the reverse transcription-polymerase chain reaction technique, we demonstrated that ZF cells express both PLD-1 and PLD-2 isozymes. Primary alcohols, which attenuate the formation of phosphatidate (the product of PLD), and cell-permeable ceramides, which inhibit PLD potently, blocked Ang-II-stimulated cortisol secretion. Furthermore, propranolol or chlorpromazine, which are potent inhibitors of phosphatidate phosphohydrolase (PAP) (the enzyme that produces diacylglycerol from phosphatidate), also blocked cortisol secretion. These data suggest that the PLD/PAP pathway plays an important role in the regulation of cortisol secretion by Ang-II in ZF cells.


Asunto(s)
Angiotensina II/farmacología , Hidrocortisona/metabolismo , Fosfolipasa D/metabolismo , Vasoconstrictores/farmacología , Zona Fascicular/efectos de los fármacos , Glándulas Suprarrenales/citología , Glándulas Suprarrenales/efectos de los fármacos , Glándulas Suprarrenales/metabolismo , Animales , Calcio/metabolismo , Bovinos , Ceramidas/metabolismo , Inhibidores Enzimáticos/farmacología , Isoenzimas/genética , Isoenzimas/metabolismo , Fosfatidato Fosfatasa/antagonistas & inhibidores , Fosfolipasa D/genética , Proteína Quinasa C/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Zona Fascicular/citología , Zona Fascicular/enzimología
19.
Mol Oncol ; 8(7): 1181-95, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24768038

RESUMEN

Sphingosine 1-phosphate (S1P) plays important roles in cell proliferation, differentiation or survival mainly through its surface G-protein-coupled receptors S1P1-5. Bone represents the major site of metastasis for prostate cancer (CaP) cells, which rely on bone-derived factors to support their proliferation and resistance to therapeutics. In the present work we have found that conditioned medium (CM) from the MC3T3 osteoblastic cell line or primary murine and human osteoblast-like cells, as well as co-culture with MC3T3 stimulate proliferation of CaP lines in S1P-dependent manner. In addition, osteoblastic-derived S1P induces resistance of CaP cells to therapeutics including chemotherapy and radiotherapy. When S1P release from osteoblastic cells is decreased (inhibition of SphK1, knock-down of SphK1 or the S1P transporter, Spns2 by siRNA) or secreted S1P neutralized with anti-S1P antibody, the proliferative and survival effects of osteoblasts on CaP cells are abolished. Because of the paracrine nature of the signaling, we studied the role of the S1P receptors expressed on CaP cells in the communication with S1P secreted by osteoblasts. Strategies aimed at down-regulating S1P1, S1P2 or S1P3 (siRNA, antagonists), established the exclusive role of the S1P/S1P1 signaling between osteoblasts and CaP cells. Bone metastases from CaP are associated with osteoblastic differentiation resulting in abnormal bone formation. We show that the autocrine S1P/S1P3 signaling is central during differentiation to mature osteoblasts by regulating Runx2 level, a key transcription factor involved in osteoblastic maturation. Importantly, differentiated osteoblasts exhibited enhanced secretion of S1P and further stimulated CaP cell proliferation in a S1P-dependent manner. By establishing the dual role of osteoblast-borne S1P on both osteoblastic differentiation and CaP cell proliferation and survival, we uncover the importance of S1P in the bone metastatic microenvironment, which may open a novel area of study for the treatment of CaP bone metastasis by targeting S1P.


Asunto(s)
Neoplasias Óseas/secundario , Lisofosfolípidos/metabolismo , Osteoblastos/patología , Próstata/patología , Neoplasias de la Próstata/patología , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/terapia , Diferenciación Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Resistencia a Antineoplásicos , Humanos , Masculino , Ratones , Osteoblastos/citología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/terapia , Transducción de Señal , Esfingosina/metabolismo
20.
Adv Cancer Res ; 117: 117-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23290779

RESUMEN

Hypoxia, defined as a poor oxygenation, has been long recognized as a hallmark of solid tumors and a negative prognostic factor for response to therapeutics and survival of patients. Cancer cells have evolved biochemical mechanisms that allow them to react and adapt to hypoxia. At the cellular level, this adaptation is under the control of two related transcription factors, HIF-1 and HIF-2 (hypoxia-inducible factor), that respond rapidly to decreased oxygen levels to activate the expression of a broad range of genes promoting neoangiogenesis, glycolysis, metastasis, increased tumor growth, and resistance to treatments. Recent studies have identified the sphingosine kinase 1/sphingosine 1-phosphate (SphK1/S1P) signaling pathway-which elicits various cellular processes including cell proliferation, cell survival, or angiogenesis-as a new regulator of HIF-1 or HIF-2 activity. In this review, we will focus on how the inhibition/neutralization of the SphK1/S1P signaling could be exploited for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Hipoxia , Lisofosfolípidos/metabolismo , Neoplasias/patología , Esfingosina/análogos & derivados , Animales , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neovascularización Patológica , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Transducción de Señal , Esfingosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA