Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4557, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38811530

RESUMEN

Glucocorticoid (GC) resistance in childhood relapsed B-cell acute lymphoblastic leukemia (B-ALL) represents an important challenge. Despite decades of clinical use, the mechanisms underlying resistance remain poorly understood. Here, we report that in B-ALL, GC paradoxically induce their own resistance by activating a phospholipase C (PLC)-mediated cell survival pathway through the chemokine receptor, CXCR4. We identify PLC as aberrantly activated in GC-resistant B-ALL and its inhibition is able to induce cell death by compromising several transcriptional programs. Mechanistically, dexamethasone (Dex) provokes CXCR4 signaling, resulting in the activation of PLC-dependent Ca2+ and protein kinase C signaling pathways, which curtail anticancer activity. Treatment with a CXCR4 antagonist or a PLC inhibitor improves survival of Dex-treated NSG mice in vivo. CXCR4/PLC axis inhibition significantly reverses Dex resistance in B-ALL cell lines (in vitro and in vivo) and cells from Dex resistant ALL patients. Our study identifies how activation of the PLC signalosome in B-ALL by Dex limits the upfront efficacy of this chemotherapeutic agent.


Asunto(s)
Dexametasona , Resistencia a Antineoplásicos , Glucocorticoides , Receptores CXCR4 , Transducción de Señal , Fosfolipasas de Tipo C , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Humanos , Animales , Transducción de Señal/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Dexametasona/farmacología , Fosfolipasas de Tipo C/metabolismo , Línea Celular Tumoral , Glucocorticoides/farmacología , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Ratones Endogámicos NOD , Supervivencia Celular/efectos de los fármacos
2.
Eur J Pharmacol ; 971: 176515, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38547958

RESUMEN

Orai1 channel capacity to control store-operated Ca2+ entry (SOCE) and B-cell functions is poorly understood and more specifically in B-cell cancers, including human lymphoma and leukemia. As compared to normal B-cells, Orai1 is overexpressed in B-chronic lymphocytic leukemia (B-CLL) and contributes in resting B-CLL to mediate an elevated basal Ca2+ level through a constitutive Ca2+ entry, and in BCR-activated B-cell to regulate the Ca2+ signaling response. Such observations were confirmed in human B-cell lymphoma and leukemia lines, including RAMOS, JOK-1, MEC-1 and JVM-3 cells. Next, the use of pharmacological Orai1 inhibitors (GSK-7975 A and Synta66) blocks constitutive Ca2+ entry and in turn affects B-cell cancer (primary and cell lines) survival and migration, controls cell cycle, and induces apoptosis through a mitochondrial and caspase-3 independent pathway. Finally, the added value of Orai1 inhibitors in combination with B-CLL drugs (ibrutinib, idelalisib, rituximab, and venetoclax) on B-CLL survival was tested, showing an additive/synergistic effect including in the B-cell cancer lines. To conclude, this study highlights the pathophysiological role of the Ca2+ channel Orai1 in B-cell cancers, and pave the way for the use of ORAI1 modulators as a plausible therapeutic strategy.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Señalización del Calcio , Supervivencia Celular , Linfocitos B/metabolismo , Línea Celular , Proteína ORAI1/metabolismo , Calcio/metabolismo , Molécula de Interacción Estromal 1/metabolismo
3.
Biochem Pharmacol ; 219: 115955, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38040093

RESUMEN

In non-excitable cells, Orai proteins represent the main channel for Store-Operated Calcium Entry (SOCE), and also mediate various store-independent Calcium Entry (SICE) pathways. Deregulation of these pathways contribute to increased tumor cell proliferation, migration, metastasis, and angiogenesis. Among Orais, Orai1 is an attractive therapeutic target explaining the development of specific modulators. Therapeutic trials using Orai1 channel inhibitors have been evaluated for treating diverse diseases such as psoriasis and acute pancreatitis, and emerging data suggest that Orai1 channel modulators may be beneficial for cancer treatment. This review discusses herein the importance of Orai1 channel modulators as potential therapeutic tools and the added value of these modulators for treating cancer.


Asunto(s)
Neoplasias , Pancreatitis , Humanos , Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Enfermedad Aguda , Neoplasias/tratamiento farmacológico , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/metabolismo
4.
Cancers (Basel) ; 15(10)2023 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-37345151

RESUMEN

Known as a key effector in relapse of acute lymphoblastic leukemia (ALL), resistance to drug-induced apoptosis, is tightly considered one of the main prognostic factors for the disease. ALL cells are constantly developing cellular strategies to survive and resist therapeutic drugs. Glucocorticoids (GCs) are one of the most important agents used in the treatment of ALL due to their ability to induce cell death. The mechanisms of GC resistance of ALL cells are largely unknown and intense research is currently focused on this topic. Such resistance can involve different cellular and molecular mechanisms, including the modulation of signaling pathways involved in the regulation of proliferation, apoptosis, autophagy, metabolism, epigenetic modifications and tumor suppressors. Recently, several studies point to the paradoxical role of GCs in many survival processes that may lead to therapy-induced resistance in ALL cells, which we called "paradoxical corticosensitivity". In this review, we aim to summarize all findings on cell survival pathways paradoxically activated by GCs with an emphasis on previous and current knowledge on gene expression and signaling pathways.

6.
Rev Physiol Biochem Pharmacol ; 183: 157-176, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-32767122

RESUMEN

The intracellular Ca2+ concentration is mainly controlled by Ca2+ channels. These channels form complexes with K+ channels, which function to amplify Ca2+ flux. In cancer cells, voltage-gated/voltage-dependent Ca2+ channels and non-voltage-gated/voltage-independent Ca2+ channels have been reported to interact with K+ channels such as Ca2+-activated K+ channels and voltage-gated K+ channels. These channels are activated by an increase in cytosolic Ca2+ concentration or by membrane depolarization, which induces membrane hyperpolarization, increasing the driving force for Ca2+ flux. These complexes, composed of K+ and Ca2+ channels, are regulated by several molecules including lipids (ether lipids and cholesterol), proteins (e.g. STIM), receptors (e.g. S1R/SIGMAR1), and peptides (e.g. LL-37) and can be targeted by monoclonal antibodies, making them novel targets for cancer research.


Asunto(s)
Neoplasias , Canales de Potasio con Entrada de Voltaje , Calcio/metabolismo , Canales de Calcio/metabolismo , Humanos , Lípidos , Neoplasias/tratamiento farmacológico , Potasio/metabolismo , Canales de Potasio/metabolismo
7.
Nutrients ; 10(9)2018 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-30200577

RESUMEN

The origin of spontaneous preference for dietary lipids in humans and rodents is debated, though recent compelling evidence has shown the existence of fat taste that might be considered a sixth taste quality. We investigated the implication of gustatory and reward brain circuits, triggered by linoleic acid (LA), a long-chain fatty acid. The LA was applied onto the circumvallate papillae for 30 min in conscious C57BL/6J mice, and neuronal activation was assessed using c-Fos immunohistochemistry. By using real-time reverse transcription polymerase chain reaction (RT-qPCR), we also studied the expression of mRNA encoding brain-derived neurotrophic factor (BDNF), Zif-268, and Glut-1 in some brain areas of these animals. LA induced a significant increase in c-Fos expression in the nucleus of solitary tract (NST), parabrachial nucleus (PBN), and ventroposterior medialis parvocellularis (VPMPC) of the thalamus, which are the regions known to be activated by gustatory signals. LA also triggered c-Fos expression in the central amygdala and ventral tegmental area (VTA), involved in food reward, in conjunction with emotional traits. Interestingly, we noticed a high expression of BDNF, Zif-268, and Glut-1 mRNA in the arcuate nucleus (Arc) and hippocampus (Hipp), where neuronal activation leads to memory formation. Our study demonstrates that oral lipid taste perception might trigger the activation of canonical gustatory and reward pathways.


Asunto(s)
Conducta Animal , Encéfalo/fisiología , Ácido Linoleico/administración & dosificación , Recompensa , Papilas Gustativas/fisiología , Percepción del Gusto , Gusto , Animales , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Ácido Linoleico/metabolismo , Masculino , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología , Plasticidad Neuronal , Proteínas Proto-Oncogénicas c-fos/metabolismo , Papilas Gustativas/metabolismo
8.
Oncogene ; 37(36): 4979-4993, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29795329

RESUMEN

Recent studies have suggested that the lipid-lowering agent simvastatin holds great promise as a cancer therapeutic; it inhibits the growth of multiple tumors, including triple-negative breast cancer. Doxorubicin- and simvastatin-induced cytotoxicity has been associated with the modulation of Ca2+ signaling, but the underlying mechanisms remain incompletely understood. Here we identify how Ca2+ signaling regulates the breast tumor cell response to doxorubicin and simvastatin. These two drugs inhibit cell survival while increasing apoptosis in two human breast cancer cell lines and five primary breast tumor specimens through the modulation of Ca2+ signaling. Signal transduction and functional studies revealed that both simvastatin and doxorubicin trigger persistent cytosolic Ca2+ release, thereby stimulating the proapoptotic BIM pathway and mitochondrial Ca2+ overload, which are responsible for metabolic dysfunction and apoptosis induction. Simvastatin and doxorubicin suppress the prosurvival ERK1/2 pathway in a Ca2+-independent and Ca2+-dependent manner, respectively. In addition, reduction of the Ca2+ signal by chelation or pharmacological inhibition significantly prevents drug-mediated anticancer signaling. Unexpectedly, a scratch-wound assay indicated that these two drugs induce rapid cell migration, while inhibiting cell invasion and colony formation in a Ca2+-dependent manner. Further, the in vivo data for MDA-MB-231 xenografts demonstrate that upon chelation of Ca2+, the ability of both drugs to reduce the tumor burden was significantly reduced via caspase-3 deactivation. Our results establish a calcium-based mechanism as crucial for executing the cell death process triggered by simvastatin and doxorubicin, and suggest that combining simvastatin with doxorubicin may be an effective regimen for the treatment of breast cancer.


Asunto(s)
Calcio/metabolismo , Doxorrubicina/farmacología , Transducción de Señal/fisiología , Simvastatina/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células MCF-7 , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Transducción de Señal/efectos de los fármacos
9.
Oncotarget ; 8(16): 27339-27352, 2017 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-28423696

RESUMEN

Previous studies have demonstrated that glucocorticoid hormones, including dexamethasone, induced alterations in intracellular calcium homeostasis in acute lymphoblastic leukemia (ALL) cells. However, the mechanism by which intracellular calcium homeostasis participates in dexamethasone sensitivity and resistance on ALL cells remains elusive. Here, we found that treatment of cells with dexamethasone resulted in increased intracellular calcium concentrations through store-operated calcium entry stimulation, which was curtailed by store-operated calcium channel blockers. We show that BAPTA-AM, an intracellular Ca2+ chelator, synergistically enhances dexamethasone lethality in two human ALL cell lines and in three primary specimens. This effect correlated with the inhibition of the prosurvival kinase ERK1/2 signaling pathway. Chelating intracellular calcium with Bapta-AM or inhibiting ERK1/2 with PD98059 significantly potentiated dexamethasone-induced mitochondrial membrane potential collapse, reactive oxygen species production, cytochrome c release, caspase-3 activity, and cell death. Moreover, we show that thapsigargin elevates intracellular free calcium ion level, and activates ERK1/2 signaling, resulting in the inhibition of dexamethasone-induced ALL cells apoptosis. Together, these results indicate that calcium-related ERK1/2 signaling pathway contributes to protect cells from dexamethasone sensitivity by limiting mitochondrial apoptotic pathway. This report provides a novel resistance pathway underlying the regulatory effect of dexamethasone on ALL cells.


Asunto(s)
Quelantes del Calcio/farmacología , Calcio/metabolismo , Dexametasona/farmacología , Resistencia a Antineoplásicos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Apoptosis/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo
11.
FASEB J ; 30(10): 3489-3500, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27358389

RESUMEN

Obesity is a major public health problem. An in-depth knowledge of the molecular mechanisms of oro-sensory detection of dietary lipids may help fight it. Humans and rodents can detect fatty acids via lipido-receptors, such as CD36 and GPR120. We studied the implication of the MAPK pathways, in particular, ERK1/2, in the gustatory detection of fatty acids. Linoleic acid, a dietary fatty acid, induced via CD36 the phosphorylation of MEK1/2-ERK1/2-ETS-like transcription factor-1 cascade, which requires Fyn-Src kinase and lipid rafts in human taste bud cells (TBCs). ERK1/2 cascade was activated by Ca2+ signaling via opening of the calcium-homeostasis modulator-1 (CALHM1) channel. Furthermore, fatty acid-evoked Ca2+ signaling and ERK1/2 phosphorylation were decreased in both human TBCs after small interfering RNA knockdown of CALHM1 channel and in TBCs from Calhm1-/- mice. Targeted knockdown of ERK1/2 by small interfering RNA or PD0325901 (MEK1/2 inhibitor) in the tongue and genetic ablation of Erk1 or Calhm1 genes impaired preference for dietary fat in mice. Lingual inhibition of ERK1/2 in healthy volunteers also decreased orogustatory sensitivity for linoleic acid. Our data demonstrate that ERK1/2-MAPK cascade is regulated by the opening of CALHM1 Ca2+ channel in TBCs to modulate orogustatory detection of dietary lipids in mice and humans.-Subramaniam, S., Ozdener, M. H., Abdoul-Azize, S., Saito, K., Malik, B., Maquart, G., Hashimoto, T., Marambaud, P., Aribi, M., Tordoff, M. G., Besnard, P., Khan, N. A. ERK1/2 activation in human taste bud cells regulates fatty acid signaling and gustatory perception of fat in mice and humans.


Asunto(s)
Ácidos Grasos/genética , Sistema de Señalización de MAP Quinasas , Papilas Gustativas/efectos de los fármacos , Gusto/efectos de los fármacos , Animales , Benzamidas/farmacología , Señalización del Calcio/efectos de los fármacos , Grasas de la Dieta/metabolismo , Difenilamina/análogos & derivados , Difenilamina/farmacología , Ácidos Grasos/metabolismo , Preferencias Alimentarias/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Ratones Noqueados , MicroARNs/genética , Obesidad/metabolismo , Gusto/fisiología , Percepción del Gusto/efectos de los fármacos , Percepción del Gusto/genética
12.
Eur J Pharmacol ; 784: 90-8, 2016 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-27179991

RESUMEN

Dexamethasone (Dex) is used as a chemotherapeutic drug in the treatment of acute lymphoblastic leukemia (ALL) because of its capacity to induce apoptosis. However, some ALL patients acquire resistance to glucocorticoids (GC). Thus, it is important to explore new agents to overcome GC resistance. The aim of the present work was to assess the ability of Pyr3, a selective inhibitor of transient receptor potential canonical 3 (TRPC3), to sensitize human ALL cells to Dex. We show here, for the first time, that Pyr3 enhances Dex sensitivity through the distraction of Dex-mediated Ca(2+) signaling in ALL cells (in vitro) and primary blasts (ex vivo) associated with mitochondrial-mediated reactive oxygen species production in ALL cells. Pyr3 alone induced Ca(2+) signaling via only endoplasmic reticulum-released Ca(2+) and exerted inhibitory effect on store-operated Ca(2+) entry in dose-dependent manner in ALL cell lines. Pre-incubation of cells with Pyr3 significantly curtailed the thapsigargin- and Dex-evoked Ca(2+) signaling in ALL cell lines. Pyr3 synergistically potentiated Dex lethality, as shown by the induction of cell mortality, G2/M cell cycle arrest and apoptosis in ALL cell lines. Moreover, Pyr3 disrupted Dex-mediated Ca(2+) signaling and increased the sensitivity of Dex-induced cell death in primary blasts from ALL patients. Additional analysis showed that co-treatment with Dex and Pyr3 results in mitochondrial membrane potential depolarization and reactive oxygen species production in ALL cells. Together, Pyr3 exhibited potential therapeutic benefit in combination with Dex to inverse glucocorticoid resistance in human ALL and probably in other lymphoid malignancies.


Asunto(s)
Apoptosis/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Dexametasona/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pirazoles/farmacología , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Humanos , Canales Catiónicos TRPC/antagonistas & inhibidores
13.
J Nutr Metab ; 2016: 2867470, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28053781

RESUMEN

Zizyphus lotus, belonging to the Rhamnaceae family, is a deciduous shrub which generally grows in arid and semiarid regions of the globe. In traditional medicine, Z. lotus is used as antidiabetes, sedative, bronchitis, and antidiarrhea by local populations. Recently, several scientific reports for health benefit and nutritional potential of bioactive compounds from this jujube have been reported. This plant is rich in polyphenols, cyclopeptide alkaloids, dammarane saponins, vitamins, minerals, amino acids, and polyunsaturated fatty acids. These identified compounds were supposed to be responsible for most of Z. lotus biologically relevant activities including antimicrobial, anti-inflammatory, hypoglycemic, antioxidant, and immunomodulatory effects. The aim of the present review was to give particular emphasis on the most recent findings on biological effects of the major groups of Zizyphus lotus components and their medical interest, notably for human nutrition, health benefit, and therapeutic impacts.

14.
BMC Complement Altern Med ; 15: 426, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26627682

RESUMEN

BACKGROUND: Pearl millet (PM), i.e., Pennisetum glaucum, is widely grown in Africa and known for its anti-oxidant and anti-hyperlipidemic properties. METHODS: The P. glaucum grains were obtained from the region of Ouled Aïssa (South of Algeria). We assessed the effects of phenolic compounds and lipids, extracted from seeds of P. glaucum, on rat lymphocyte proliferation, activated by phorbol 12-myristate 13-acetate and ionomycin. In order to explore signaling pathway, triggered by these compounds, we assessed interleukin-2 (IL-2) mRNA expression and extracellular signal-regulated kinase-1/2 (ERK1/ERK2) phosphorylation. Finally, we determined increases in free intracellular Ca(2+) concentrations, [Ca(2+)]i, by employing Fura-2/AM in rat lymphocytes. RESULTS: The composition of P. glaucum grains in polyphenols was estimated to be 1660 µg gallic acid equivalents (GAE)/g. Lipids represented 4.5 %, and more than 72% of the fatty acids belonged to unsaturated family. Our investigation showed that both lipid and phenolic compounds inhibited mitogen-induced T-cell proliferation. Compared with phenolic compounds, lipids exerted weaker effects on ERK-1/ERK2 phosphorylation and Ca(2+) signaling in mitogen-activated T-cells. CONCLUSION: We conclude that the immunomodulatory effects of P. glaucum could be contributed by its phenolic and lipid contents.


Asunto(s)
Calcio/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Lípidos/farmacología , Pennisetum , Polifenoles/farmacología , Linfocitos T/efectos de los fármacos , Animales , Antioxidantes/farmacología , Proliferación Celular/efectos de los fármacos , Grano Comestible/química , Hipolipemiantes/farmacología , Activación de Linfocitos/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Pennisetum/química , Extractos Vegetales/farmacología , Ratas , Semillas , Transducción de Señal , Linfocitos T/inmunología
15.
Biochimie ; 109: 60-6, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25528298

RESUMEN

The desert gerbil, Psammomys obesus, is a unique polygenic animal model of metabolic syndrome (insulin resistance, obesity and type 2 diabetes), and these pathological conditions resemble to those in human beings. In this study, the animals were fed ad libitum either a natural diet (ND) which contained desertic halophile plants or a standard laboratory diet (STD) or a diet which contained eicosapentaenoic acid (EPA), hence, termed as EPA diet (EPAD). In EPAD, 50% of total lipid content was replaced by EPA oil. By employing real-time PCR, we assessed liver expression of key genes involved in fatty acid metabolism such as PPAR-α, SREBP-1c, LXR-α and CHREBP. We also studied the expression of two inflammatory genes, i.e., TNF-α and IL-1ß, in liver and adipose tissue of these animals. The STD, considered to be a high caloric diet for this animal, triggered insulin resistance and high lipid levels, along with high hepatic SREBP-1c, LXR-α and CHREBP mRNA expression. TNF-α and IL-1ß mRNA were also high in liver of STD fed animals. Feeding EPAD improved plasma glucose, insulin and triacylglycerol levels along with hepatic lipid composition. These observations suggest that EPA exerts beneficial effects in P. obesus.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Ácido Eicosapentaenoico/farmacología , Ácidos Grasos/metabolismo , Mediadores de Inflamación/metabolismo , Hígado/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Gerbillinae , Insulina/sangre , Interleucina-1beta/genética , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Receptores X del Hígado , Masculino , Receptores Nucleares Huérfanos/genética , PPAR alfa/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Triglicéridos/sangre , Factor de Necrosis Tumoral alfa/genética
16.
Biochimie ; 96: 8-13, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23774298

RESUMEN

Recent compelling evidences from rodent and human studies raise the possibility for an additional sixth taste modality devoted to oro-gustatory perception of dietary lipids. Understanding the mechanisms underlying oro-gustatory detection of dietary fat is critical for the prevention and treatment of obesity. A number of studies have suggested that lingual CD36, a glycoprotein, highly expressed by circumvallate papillae of the tongue, is implicated in the perception of dietary fat taste. G protein-coupled receptors (GPCRs) are important signaling molecules for many aspects of cellular functions. It has been shown that these receptors, particularly GPR120, are also involved in lipid taste perception. We have shown that dietary long-chain fatty acids (LCFAs), in CD36-positive taste bud cells (TBC), induce increases in free intracellular Ca(2+) concentrations, [Ca(2+)]i, by recruiting Ca(2+) from endoplasmic reticulum (ER) pool via inositol 1,4,5-triphosphate production, followed by Ca(2+) influx via opening of store-operated Ca(2+) (SOC) channels. GPR120 is also coupled to increases in [Ca(2+)]i by dietary fatty acids. We observed that stromal interaction molecule 1 (STIM1), a sensor of Ca(2+) depletion in the ER, mediated fatty acid-induced Ca(2+) signaling and spontaneous preference for fat in the mouse. In this review article, we discuss the recent advances and unresolved roles of CD36 and GPR120 in lipid taste signaling in taste bud cells.


Asunto(s)
Antígenos CD36/fisiología , Señalización del Calcio , Grasas de la Dieta/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Papilas Gustativas/metabolismo , Animales , Humanos , Gusto , Papilas Gustativas/citología
17.
PLoS One ; 8(8): e68532, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936306

RESUMEN

Since the increasing prevalence of obesity is one of the major health problems of the modern era, understanding the mechanisms of oro-gustatory detection of dietary fat is critical for the prevention and treatment of obesity. We have conducted the present study on Psammomys obesus, the rodent desert gerbil which is a unique polygenic natural animal model of obesity. Our results show that obese animals exhibit a strong preference for lipid solutions in a two-bottle test. Interestingly, the expression of CD36, a lipido-receptor, in taste buds cells (TBC), isolated from circumvallate papillae, was decreased at mRNA level, but remained unaltered at protein level, in obese animals. We further studied the effects of linoleic acid (LA), a long-chain fatty acid, on the increases in free intracellular calcium (Ca(2+)) concentrations, [Ca(2+)]i, in the TBC of P. obesus. LA induced increases in [Ca(2+)]i, largely via CD36, from intracellular pool, followed by the opening of store-operated Ca(2+) (SOC) channels in the TBC of these animals. The action of this fatty acid on the increases in [Ca(2+)]i was higher in obese animals than that in controls. However, the release of Ca(2+) from intracellular stores, studied also by employing thapsigargin, was lower in TBC of obese animals than control rodents. In this study, we show, for the first time, that increased lipid intake and altered Ca(2+) signaling in TBC are associated with obesity in Psammomys obesus.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Grasas de la Dieta/farmacología , Obesidad/patología , Obesidad/fisiopatología , Papilas Gustativas/patología , Percepción del Gusto/efectos de los fármacos , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Susceptibilidad a Enfermedades , Preferencias Alimentarias/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Gerbillinae , Ácido Linoleico/farmacología , Masculino , Obesidad/metabolismo , Papilas Gustativas/efectos de los fármacos , Papilas Gustativas/metabolismo , Papilas Gustativas/fisiopatología , Tapsigargina/farmacología
18.
J Lipid Res ; 54(9): 2485-94, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23840049

RESUMEN

A relationship between orosensory detection of dietary lipids, regulation of fat intake, and body mass index was recently suggested. However, involved mechanisms are poorly understood. Moreover, whether obesity can directly modulate preference for fatty foods remains unknown. To address this question, exploration of the oral lipid sensing system was undertaken in diet-induced obese (DIO) mice. By using a combination of biochemical, physiological, and behavioral approaches, we found that i) the attraction for lipids is decreased in obese mice, ii) this behavioral change has an orosensory origin, iii) it is reversed in calorie-restricted DIO mice, revealing an inverse correlation between fat preference and adipose tissue size, iv) obesity suppresses the lipid-mediated downregulation of the lipid-sensor CD36 in circumvallate papillae, usually found during the refeeding of lean mice, and v) the CD36-dependent signaling cascade controlling the intracellular calcium levels ([Ca(2+)]i) in taste bud cells is decreased in obese mice. Therefore, obesity alters the lipid-sensing system responsible for the oral perception of dietary lipids. This phenomenon seems to take place through a CD36-mediated mechanism, leading to changes in eating behavior.


Asunto(s)
Antígenos CD36/metabolismo , Grasas de la Dieta/farmacología , Obesidad/fisiopatología , Percepción del Gusto/efectos de los fármacos , Lengua/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Animales , Conducta Animal , Señalización del Calcio/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Preferencias Alimentarias/efectos de los fármacos , Preferencias Alimentarias/fisiología , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , Obesidad/psicología , Lengua/citología , Lengua/efectos de los fármacos
19.
Int Immunopharmacol ; 15(2): 364-71, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23219580

RESUMEN

We assessed the effects of Zizyphus lotus L. (Desf.) polyphenols (ZLP) on T-cell signaling and proliferation. Our results showed that ZLP exerted no effect on the increases in intracellular free calcium concentrations, [Ca(2+)]i, in human Jurkat T-cells. However, ZLP modulated the thapsigargin-induced increases in [Ca(2+)]i in these cells. ZLP treatment was found to decrease the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2). In addition, ZLP induced a rapid (t1/2=33s) and dose-dependent decrease in intracellular pH (pHi) in human Jurkat T-cells. Furthermore, ZLP significantly curtailed T-cell proliferation by diminishing their progression from S to G2/M phase of cell cycle, and the expression of interleukin-2 (IL-2) mRNA. Taken together, the results of the present study demonstrate that ZLP modulate cell signaling and exert immunosuppressive effects in human T-cells.


Asunto(s)
Terapia de Inmunosupresión , Inflamación/inmunología , Interleucina-2/metabolismo , Polifenoles/farmacología , Linfocitos T/efectos de los fármacos , Ziziphus/química , Señalización del Calcio/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Frutas/química , Regulación de la Expresión Génica , Humanos , Interleucina-2/genética , Células Jurkat , ARN Mensajero/análisis , Linfocitos T/inmunología , Tapsigargina/inmunología
20.
J Clin Invest ; 122(6): 2267-82, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22546859

RESUMEN

Understanding the mechanisms underlying oro-gustatory detection of dietary fat is critical for the prevention and treatment of obesity. The lipid-binding glycoprotein CD36, which is expressed by circumvallate papillae (CVP) of the mouse tongue, has been implicated in oro-gustatory perception of dietary lipids. Here, we demonstrate that stromal interaction molecule 1 (STIM1), a sensor of Ca(2+) depletion in the endoplasmic reticulum, mediates fatty acid-induced Ca(2+) signaling in the mouse tongue and fat preference. We showed that linoleic acid (LA) induced the production of arachidonic acid (AA) and lysophosphatidylcholine (Lyso-PC) by activating multiple phospholipase A2 isoforms via CD36. This activation triggered Ca(2+) influx in CD36-positive taste bud cells (TBCs) purified from mouse CVP. LA also induced the production of Ca(2+) influx factor (CIF). STIM1 was found to regulate LA-induced CIF production and the opening of multiple store-operated Ca(2+) (SOC) channels. Furthermore, CD36-positive TBCs from Stim1-/- mice failed to release serotonin, and Stim1-/- mice lost the spontaneous preference for fat that was observed in wild-type animals. Our results suggest that fatty acid-induced Ca(2+) signaling, regulated by STIM1 via CD36, might be implicated in oro-gustatory perception of dietary lipids and the spontaneous preference for fat.


Asunto(s)
Antígenos CD36/metabolismo , Señalización del Calcio/fisiología , Grasas de la Dieta/farmacología , Preferencias Alimentarias/fisiología , Glicoproteínas de Membrana/metabolismo , Papilas Gustativas/metabolismo , Percepción del Gusto/fisiología , Animales , Ácido Araquidónico/genética , Ácido Araquidónico/metabolismo , Antígenos CD36/genética , Canales de Calcio/genética , Canales de Calcio/metabolismo , Ácido Linoleico/farmacología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Serotonina/genética , Serotonina/metabolismo , Molécula de Interacción Estromal 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...