Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 50
Filtrer
1.
Biomolecules ; 14(4)2024 Mar 27.
Article de Anglais | MEDLINE | ID: mdl-38672424

RÉSUMÉ

Originally developed as a chemotherapeutic agent, miltefosine (hexadecylphosphocholine) is an inhibitor of phosphatidylcholine synthesis with proven antiparasitic effects. It is the only oral drug approved for the treatment of Leishmaniasis and American Trypanosomiasis (Chagas disease). Although its precise mechanisms are not yet fully understood, miltefosine exhibits broad-spectrum anti-parasitic effects primarily by disrupting the intracellular Ca2+ homeostasis of the parasites while sparing the human hosts. In addition to its inhibitory effects on phosphatidylcholine synthesis and cytochrome c oxidase, miltefosine has been found to affect the unique giant mitochondria and the acidocalcisomes of parasites. Both of these crucial organelles are involved in Ca2+ regulation. Furthermore, miltefosine has the ability to activate a specific parasite Ca2+ channel that responds to sphingosine, which is different to its L-type VGCC human ortholog. Here, we aimed to provide an overview of recent advancements of the anti-parasitic mechanisms of miltefosine. We also explored its multiple molecular targets and investigated how its pleiotropic effects translate into a rational therapeutic approach for patients afflicted by Leishmaniasis and American Trypanosomiasis. Notably, miltefosine's therapeutic effect extends beyond its impact on the parasite to also positively affect the host's immune system. These findings enhance our understanding on its multi-targeted mechanism of action. Overall, this review sheds light on the intricate molecular actions of miltefosine, highlighting its potential as a promising therapeutic option against these debilitating parasitic diseases.


Sujet(s)
Calcium , Maladie de Chagas , Homéostasie , Leishmaniose , Phosphoryl-choline , Phosphoryl-choline/analogues et dérivés , Humains , Phosphoryl-choline/pharmacologie , Phosphoryl-choline/usage thérapeutique , Maladie de Chagas/traitement médicamenteux , Maladie de Chagas/parasitologie , Maladie de Chagas/métabolisme , Calcium/métabolisme , Leishmaniose/traitement médicamenteux , Leishmaniose/métabolisme , Leishmaniose/parasitologie , Homéostasie/effets des médicaments et des substances chimiques , Animaux , Antiprotozoaires/pharmacologie , Antiprotozoaires/usage thérapeutique , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Leishmania/effets des médicaments et des substances chimiques , Leishmania/métabolisme , Trypanosoma cruzi/effets des médicaments et des substances chimiques , Trypanosoma cruzi/métabolisme
2.
Parasitol Res ; 123(3): 166, 2024 Mar 20.
Article de Anglais | MEDLINE | ID: mdl-38506929

RÉSUMÉ

The hemoparasite Trypanosoma equiperdum belongs to the Trypanozoon subgenus and includes several species that are pathogenic to animals and humans in tropical and subtropical areas across the world. As with all eukaryotic organisms, Ca2+ is essential for these parasites to perform cellular processes thus ensuring their survival across their life cycle. Despite the established paradigm to study proteins related to Ca2+ homeostasis as potential drug targets, so far little is known about Ca2+ entry into trypanosomes. Therefore, in the present study, the presence of a plasma membrane Ca2+-channel in T. equiperdum (TeCC), activated by sphingosine and inhibited by verapamil, is described. The TeCC was cloned and analyzed using bioinformatic resources, which confirmed the presence of several domains, motifs, and a topology similar to the Ca2+ channels found in higher eukaryotes. Biochemical and confocal microscopy assays using antibodies raised against an internal region of human L-type Ca2+ channels indicate the presence of a protein with similar predicted molar mass to the sequence analyzed, located at the plasma membrane of T. equiperdum. Physiological assays based on Fura-2 signals and Mn2+ quenching performed on whole parasites showed a unidirectional Ca2+ entry, which is activated by sphingosine and blocked by verapamil, with the distinctive feature of insensitivity to nifedipine and Bay K 8644. This suggests a second Ca2+ entry for T. equiperdum, different from the store-operated Ca2+ entry (SOCE) previously described. Moreover, the evidence presented here for the TeCC indicates molecular and pharmacological differences with their mammal counterparts, which deserve further studies to evaluate the potential of this channel as a drug target.


Sujet(s)
Sphingosine , Trypanosoma , Animaux , Humains , Sphingosine/pharmacologie , Vérapamil/pharmacologie , Membrane cellulaire/métabolisme , Calcium/métabolisme , Mammifères
3.
Bioorg Chem ; 144: 107112, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38237390

RÉSUMÉ

We report here the virtual screening design, synthesis and activity of eight new inhibitors of SphK1. For this study we used a pre-trained Graph Convolutional Network (GCN) combined with docking calculations. This exploratory analysis proposed nine compounds from which eight displayed significant inhibitory effect against sphingosine kinase 1 (SphK1) demonstrating a high level of efficacy for this approach. Four of these compounds also displayed anticancer activity against different tumor cell lines, and three of them (5), (6) and (7) have shown a wide inhibitory action against many of the cancer cell line tested, with GI50 below 5 µM, being (5) the most promising with TGI below 10 µM for the half of cell lines. Our results suggest that the three most promising compounds reported here are the pyrimidine-quinolone hybrids (1) and (6) linked by p-aminophenylsulfanyl and o-aminophenol fragments respectively, and (8) without such aryl linker. We also performed an exhaustive study about the molecular interactions that stabilize the different ligands at the binding site of SphK1. This molecular modeling analysis was carried out by using combined techniques: docking calculations, MD simulations and QTAIM analysis. In this study we also included PF543, as reference compound, in order to better understand the molecular behavior of these ligands at the binding site of SphK1.These results provide useful information for the design of new inhibitors of SphK1 possessing these structural scaffolds.


Sujet(s)
Antinéoplasiques , Phosphotransferases (Alcohol Group Acceptor) , Quinolinone , Quinolinone/pharmacologie , Inhibiteurs de protéines kinases , Antinéoplasiques/composition chimique , Modèles moléculaires , Lignée cellulaire tumorale , Simulation de docking moléculaire , Tests de criblage d'agents antitumoraux , Prolifération cellulaire , Relation structure-activité , Structure moléculaire
4.
Int J Mol Sci ; 24(23)2023 Nov 23.
Article de Anglais | MEDLINE | ID: mdl-38068956

RÉSUMÉ

The objective of this study was to investigate whether the activity of enzymes involved in sphingolipid catabolism could be biomarkers to predict early renal damage in streptozotocin (STZ)-induced diabetic rats and Angiotensin II (Ang II)-induced hypertension rats. Diabetic and hypertensive rats had no changes in plasma creatinine concentration. However, transmission electron microscopy (TEM) analysis showed slight ultrastructural changes in the glomeruli and tubular epithelial cells from diabetic and hypertensive rats. Our results show that the acid sphingomyelinase (aSMase) and neutral sphingomyelinase (nSMase) activity increased in the urine of diabetic rats and decreased in hypertensive rats. Only neutral ceramidase (nCDase) activity increased in the urine of diabetic rats. Furthermore, the immunofluorescence demonstrated positive staining for the nSMase, nCDase, and sphingosine kinase (SphK1) in glomerular mesangial cells, proximal tubule, ascending thin limb of the loop of Henle, thick ascending limb of Henle's loop, and principal cells of the collecting duct in the kidney. In conclusion, our results suggest that aSMase and nCDase activity in urine could be a novel predictor of early slight ultrastructural changes in the nephron, aSMase and nCDase as glomerular injury biomarkers, and nSMase as a tubular injury biomarker in diabetic and hypertensive rats.


Sujet(s)
Diabète expérimental , Hypertension artérielle , Rats , Animaux , Sphingomyeline phosphodiesterase/métabolisme , Diabète expérimental/métabolisme , Rein/métabolisme , Néphrons/métabolisme , Sphingolipides
5.
Rev. méd. Chile ; 151(10): 1375-1384, oct. 2023. tab, ilus
Article de Espagnol | LILACS | ID: biblio-1565656

RÉSUMÉ

Siponimod es un medicamento inmunosupresor selectivo, desarrollado como la primera terapia oral para la esclerosis múltiple secundaria progresiva activa. Este medicamento actúa modulando el receptor de esfingosina 1 fosfato (S1P), como antagonista de S1P1 y S1P5, evitando así la salida de linfocitos desde los nódulos linfáticos y previniendo procesos inflamatorios en el Sistema Nervioso Central que desencadenan una desmielinización. Existe amplio conocimiento científico respecto a que la administración del medicamento a pacientes va a depender de sus características farmacogenéticas, por lo que la FDA recomienda fuertemente realizar un estudio de genotipificación de la enzima que metaboliza siponimod, CYP2C9, cuyas variantes genéticas *2 y *3 clasifican a pacientes como metabolizadores pobres, extensivos o rápidos. Para pacientes homocigotos de CYP2C9*3 siponimod está totalmente contraindicado. Adicionalmente, antes de su prescripción se debe realizar un electrocardiograma, evaluaciones del estado de anticuerpos, oftálmica, estado de vacunación contra varicela y recuento de linfocitos periféricos, ya que el efecto del medicamento es dependiente de la dosis administrada, por lo que se realiza un proceso de titulación en dosis desde los 0,25mg hasta los 2 mg. El protocolo farmacoterapéutico de siponimod es reflejo fidedigno de la utilidad de la farmacogenética en la medicina personalizada..


Siponimod is a selective immunosuppressive medication, developed as the first oral therapy for active secondary progressive multiple sclerosis. This medication acts by modulating the sphingosine 1 phosphate (S1P) receptor, as an antagonist of S1P1 and S1P5, thus preventing the egress of lymphocytes from lymph nodes and preventing inflammatory processes in the Central Nervous System that trigger demyelination. There is extensive scientific knowledge regarding the administration of the medication to patients, which will depend on their pharmacogenetic characteristics. Therefore, the FDA strongly recommends conducting a genotyping study of the enzyme that metabolizes siponimod, CYP2C9, whose genetic variants *2 and *3 classify patients as poor, extensive, or rapid metabolizers. Siponimod is completely contraindicated for patients who are homozygous for CYP2C9*3. Additionally, before prescribing it, an electrocardiogram, assessments of antibody status, ophthalmic evaluation, varicella vaccination status, and peripheral lymphocyte count should be conducted, as the medication's effect is dose-dependent. Therefore, a titration process is carried out starting from 0.25mg up to 2 mg. The pharmacotherapeutic protocol of siponimod is a reliable reflection of the utility of pharmacogenetics in personalized medicine.


Sujet(s)
Humains , Azétidines/administration et posologie , Azétidines/usage thérapeutique , Composés benzyliques/administration et posologie , Composés benzyliques/usage thérapeutique , Sclérose en plaques chronique progressive/génétique , Sclérose en plaques chronique progressive/traitement médicamenteux , Pharmacogénétique , Relation dose-effet des médicaments , Modulateurs des récepteurs de la sphingosine 1 phosphate/administration et posologie , Modulateurs des récepteurs de la sphingosine 1 phosphate/usage thérapeutique , Immunosuppresseurs/administration et posologie , Immunosuppresseurs/usage thérapeutique
6.
Front Oncol ; 13: 1143881, 2023.
Article de Anglais | MEDLINE | ID: mdl-37020867

RÉSUMÉ

The treatment of chronic lymphocytic leukemia (CLL) patients with venetoclax-based regimens has demonstrated efficacy and a safety profile, but the emergence of resistant cells and disease progression is a current complication. Therapeutic target of sphingosine kinases (SPHK) 1 and 2 has opened new opportunities in the treatment combinations of cancer patients. We previously reported that the dual SPHK1/2 inhibitor, SKI-II enhanced the in vitro cell death triggered by fludarabine, bendamustine or ibrutinib and reduced the activation and proliferation of chronic lymphocytic leukemia (CLL) cells. Since we previously showed that autologous activated T cells from CLL patients favor the activation of CLL cells and the generation of venetoclax resistance due to the upregulation of BCL-XL and MCL-1, we here aim to determine whether SPHK inhibitors affect this process. To this aim we employed the dual SPHK1/2 inhibitor SKI-II and opaganib, a SPHK2 inhibitor that is being studied in clinical trials. We found that SPHK inhibitors reduce the activation of CLL cells and the generation of venetoclax resistance induced by activated T cells mainly due to a reduced upregulation of BCL-XL. We also found that SPHK2 expression was enhanced in CLL cells by activated T cells of the same patient and the presence of venetoclax selects resistant cells with high levels of SPHK2. Of note, SPHK inhibitors were able to re-sensitize already resistant CLL cells to a second venetoclax treatment. Our results highlight the therapeutic potential of SPHK inhibitors in combination with venetoclax as a promising treatment option for the patients.

7.
Arch. cardiol. Méx ; Arch. cardiol. Méx;93(1): 88-95, ene.-mar. 2023. tab, graf
Article de Espagnol | LILACS-Express | LILACS | ID: biblio-1429709

RÉSUMÉ

Resumen Los esfingolípidos (esfingomielina, glucolípidos y gangliósidos) se localizan en las membranas celulares, el plasma y las lipoproteínas. En pacientes con enfermedades cardiovasculares, renales y metabólicas, el perfil de los esfingolípidos y sus metabolitos (ceramida, esfingosina y esfingosina-1-fosfato) se modifica, y estos cambios pueden explicar las alteraciones en algunas respuestas celulares, como la apoptosis. Además, se ha sugerido que la esfingosina y la esfingosina-1-fosfato previenen la COVID-19. En esta revisión también se mencionan brevemente las técnicas que permiten el estudio de los esfingolípidos y sus metabolitos.


Abstract Sphingolipids (sphingomyelin, glycolipids, gangliosides) are located in cell membranes, plasma, and lipoproteins. In patients with cardiovascular, renal, and metabolic diseases, the profile of sphingolipids and their metabolites (ceramide, sphingosine, and sphingosine-1-phosphate) is modified, and these changes may explain the alterations in some cellular responses such as apoptosis. Furthermore, sphingosine and sphingosine-1-phosphate have been suggested to prevent COVID-19. This review also briefly mentions the techniques that allow us to study sphingolipids and their metabolites.

8.
Arch Cardiol Mex ; 93(1): 88-95, 2023.
Article de Anglais | MEDLINE | ID: mdl-36757794

RÉSUMÉ

Sphingolipids (sphingomyelin, glycolipids, gangliosides) are located in cell membranes, plasma, and lipoproteins. In patients with cardiovascular, renal, and metabolic diseases, the profile of sphingolipids and their metabolites (ceramide, sphingosine, and sphingosine-1-phosphate) is modified, and these changes may explain the alterations in some cellular responses such as apoptosis. Furthermore, sphingosine and sphingosine-1-phosphate have been suggested to prevent COVID-19. This review also briefly mentions the techniques that allow us to study sphingolipids and their metabolites.


Los esfingolípidos (esfingomielina, glucolípidos y gangliósidos) se localizan en las membranas celulares, el plasma y las lipoproteínas. En pacientes con enfermedades cardiovasculares, renales y metabólicas, el perfil de los esfingolípidos y sus metabolitos (ceramida, esfingosina y esfingosina-1-fosfato) se modifica, y estos cambios pueden explicar las alteraciones en algunas respuestas celulares, como la apoptosis. Además, se ha sugerido que la esfingosina y la esfingosina-1-fosfato previenen la COVID-19. En esta revisión también se mencionan brevemente las técnicas que permiten el estudio de los esfingolípidos y sus metabolitos.


Sujet(s)
COVID-19 , Maladies métaboliques , Humains , Sphingosine/métabolisme , Sphingolipides/métabolisme
9.
Life Sci ; 316: 121404, 2023 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-36681184

RÉSUMÉ

Epithelial renal cells have the ability to adopt different cellular phenotypes through epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET). These processes are increasingly recognized as important repair factors following acute renal tubular injury. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid with impact on proliferation, growth, migration, and differentiation which has significant implication in various diseases including cancer and kidney fibrosis. Here we demonstrated that S1P can exert by activating S1P receptor 2 (S1PR2) different functions depending on the stage of cell differentiation. We observed that the differences in the migratory profile of Madin-Darby canine kidney (MDCK) cells depend both on their stage of cell differentiation and the activity of S1PR2, a receptor that can either promote or inhibit the migratory process. Meanwhile in non-differentiated cells S1PR2 activation avoids migration, it is essential on fully differentiated cells. This is the first time that an antagonist effect of S1PR2 was reported for the same cell type. Moreover, in fully differentiated cells, S1PR2 activation is crucial for the progression of EMT - characterized by adherent junctions disassembly, ß-catenin and SNAI2 nuclear translocation and vimentin expression- and depends on ERK 1/2 activation and nuclear translocation. These findings provide a new perspective about the different S1PR2 functions depending on the stage of cell differentiation that can be critical to the modulation of renal epithelial cell plasticity, potentially paving the way for innovative research with pathophysiologic relevance.


Sujet(s)
Différenciation cellulaire , Rein , Récepteurs de la sphingosine-1-phosphate , Animaux , Chiens , Lysophospholipides/métabolisme , Cellules rénales canines Madin-Darby , Récepteurs aux lysosphingolipides/métabolisme , Rein/cytologie
10.
Brain Res ; 1799: 148171, 2023 01 15.
Article de Anglais | MEDLINE | ID: mdl-36410428

RÉSUMÉ

Sphingolipid-1-phosphate (S1P) signaling through the activation S1P receptors (S1PRs) plays critical roles in cellular events in the brain. Aberrant S1P metabolism has been identified in the brains of Alzheimer's disease (AD) patients. Our recent studies have shown that treatment with fingolimod, an analog of sphingosine, provides neuroprotective effects in five familiar Alzheimer disease (5xFAD) transgenic mice, resulting in the reduction of amyloid-ß (Aß) neurotoxicity, inhibition of activation of microglia and astrocytes, increased hippocampal neurogenesis, and improved learning and memory. However, the pathways by which dysfunctional S1P and S1PR signaling may associate with the development of AD-like pathology remain unknown. In this study, we investigated the alteration of signaling of S1P/S1P receptor 1 (S1PR1), the most abundant S1PR subtype in the brain, in the cortex of 5xFAD transgenic mice at 3, 8, and 14 months of age. Compared to non-transgenic wildtype (WT) littermates, we found significant decreased levels of sphingosine kinases (SphKs), increased S1P lyase (S1PL), and increased S1PR1 in 8- and 14-month-old, but not in 3-month-old 5xFAD mice. Furthermore, we detected increased activation of the S1PR1 downstream pathway of Akt/mTor/Tau signaling in aging 5xFAD mice. Treatment with fingolimod from 1 to 8 months of age reversed the levels of SphKs, S1PL, and furthermore, those of S1PR1 and its downstream pathway of Akt/mTor/Tau signaling. Together the data reveal that dysregulation of S1P and S1PR signaling may associate with the development of AD-like pathology through Akt/mTor/Tau signaling.


Sujet(s)
Maladie d'Alzheimer , Sphingosine , Souris , Animaux , Chlorhydrate de fingolimod/pharmacologie , Maladie d'Alzheimer/métabolisme , Récepteurs de la sphingosine-1-phosphate , Protéines proto-oncogènes c-akt , Lysophospholipides/métabolisme , Modèles animaux de maladie humaine , Souris transgéniques , Sérine-thréonine kinases TOR
11.
Odovtos (En línea) ; 24(1)abr. 2022.
Article de Anglais | LILACS, SaludCR | ID: biblio-1386575

RÉSUMÉ

Abstract The aim of this study was the quantification of Sphingosine-1-phosphate (S1P) in periodontal pockets of patients with periodontitis. This is an observational, descriptive, case-control study. Thirty subjects were selected: 15 controls and 15 cases. A periodontal study was conducted following the parameters of AAP 2017 for the diagnosis of periodontal diseases. A sample of saliva and gingival crevicular fluid was obtained from each subject and then analyzed with the Human S1P Elisa kit (MyBioSource #MBS2516132) accordingly to the manufacturer's instructions, in order to verify the presence of S1P and quantify it´s concentration when founded. Results showed a significant difference (p=0.05) between cases and controls. In the case of saliva samples, the concentration of S1P was higher than the ones found in the control group (72.94 ng/mL and 45.12 ng/mL). For GCF, a higher amount of S1P was found in patients with POD (20.09 ng/mL and 15.20 ng/mL). This work raises a possible route of bone metabolism, inflammatory process, and identification of periodontitis through oral quantification of S1P, however, future studies are needed.


Resumen El propósito de este estudio fue la cuantificación de Esfingosina-1-Fosfato (S1P) en las bolsas periodontales de pacientes con periodontitis. Estudio observacional, descriptivo de casos y controles. 30 sujetos fueron seleccionados de los cuales 15 controles y 15 casos. Se realizó un estudio periodontal completo siguiendo los parámetros establecidos por la AAP en 2017 para el diagnóstico de las enfermedades periodontales. Se tomaron muestras de saliva y de líquido crevicular gingival de cada sujeto estudiado y se analizaron con el ELISA kit humano para S1P (MyBiosource #MBS2516132) y de acuerdo con las instrucciones del fabricante, se realizó para cuantificar la presencia d S1P en las muestras estudiadas. Los resultados mostraron diferencia significativa (p=0.05) entre casos y controles. En el caso de las muestras de saliva, la concentración de S1P en controles fue mayor (72.94 ng/mL y 45.12 ng/mL). Para Líquido crevicular gingival, se encontró mayor cantidad de S1P en los pacientes con periodontitis (20.09 ng/mL y 15.20 ng/mL). Este estudio plantea una posible ruta de metabolismo óseo, proceso inflamatorio e identificación de la Periodontitis a través de la cuantificación oral de S1P, sin embargo se necesitan estudios futuros.


Sujet(s)
Humains , Parodontite , Récepteurs de la sphingosine-1-phosphate/analyse
12.
Int J Mol Sci ; 23(5)2022 Mar 07.
Article de Anglais | MEDLINE | ID: mdl-35270028

RÉSUMÉ

The profile of sphingomyelin and its metabolites shows changes in the plasma, organs, and tissues of patients with cardiovascular, renal, and metabolic diseases. The objective of this study was to investigate the effect of empagliflozin on the levels of sphingomyelin and its metabolites, as well as on the activity of acid and neutral sphingomyelinase (aSMase and nSMase) and neutral ceramidase (nCDase) in the plasma, kidney, heart, and liver of streptozotocin-induced diabetic and Angiotensin II (Ang II)-induced hypertension rats. Empagliflozin treatment decreased hyperglycemia in diabetic rats whereas blood pressure remained elevated in hypertensive rats. In diabetic rats, empagliflozin treatment decreased sphingomyelin in the plasma and liver, ceramide in the heart, sphingosine-1-phosphate (S1P) in the kidney, and nCDase activity in the plasma, heart, and liver. In hypertensive rats, empagliflozin treatment decreased sphingomyelin in the plasma, kidney, and liver; S1P in the plasma and kidney; aSMase in the heart, and nCDase activity in the plasma, kidney, and heart. Our results suggest that empagliflozin downregulates the interaction of the de novo pathway and the catabolic pathway of sphingolipid metabolism in the diabetes, whereas in Ang II-dependent hypertension, it only downregulates the sphingolipid catabolic pathway.


Sujet(s)
Diabète expérimental , Hypertension artérielle , Animaux , Composés benzhydryliques , Céramides/métabolisme , Diabète expérimental/traitement médicamenteux , Glucosides , Humains , Hypertension artérielle/traitement médicamenteux , Rats , Sphingolipides/métabolisme , Sphingomyeline phosphodiesterase/métabolisme , Sphingomyéline
13.
Cell Signal ; 87: 110123, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34438016

RÉSUMÉ

The fibrotic process could be easily defined as a pathological excess of extracellular matrix deposition, leading to disruption of tissue architecture and eventually loss of function; however, this process involves a complex network of several signal transduction pathways. Virtually almost all organs could be affected by fibrosis, the most affected are the liver, lung, skin, kidney, heart, and eyes; in all of them, the transforming growth factor-beta (TGF-ß) has a central role. The canonical and non-canonical signal pathways of TGF-ß impact the fibrotic process at the cellular and molecular levels, inducing the epithelial-mesenchymal transition (EMT) and the induction of profibrotic gene expression with the consequent increase in proteins such as alpha-smooth actin (α-SMA), fibronectin, collagen, and other extracellular matrix proteins. Recently, it has been reported that some molecules that have not been typically associated with the fibrotic process, such as nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4), mammalian target of rapamycin (mTOR), histone deacetylases (HDAC), and sphingosine-1 phosphate (S1P); are critical in its development. In this review, we describe and discuss the role of these new players of fibrosis and the convergence with TGF-ß signaling pathways, unveiling new insights into the panorama of fibrosis that could be useful for future therapeutic targets.


Sujet(s)
Sphingosine , Facteur de croissance transformant bêta , Fibrose , Amis , Histone deacetylases , Humains , NADPH Oxidase 4 , Sphingosine/métabolisme , Sérine-thréonine kinases TOR , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta-1/métabolisme
14.
Article de Anglais | MEDLINE | ID: mdl-33454434

RÉSUMÉ

Collecting duct cells are physiologically subject to the hypertonic environment of the kidney. This condition is necessary for kidney maturation and function but represents a stress condition that requires active strategies to ensure epithelial integrity. Madin-Darby Canine Kidney (MDCK) cells develop the differentiated phenotype of collecting duct cells when subject to hypertonicity, serving as a model to study epithelial preservation and homeostasis in this particular environment. The integrity of epithelia is essential to achieve the required functional barrier. One of the mechanisms that ensure integrity is cell extrusion, a process initiated by sphingosine-1-phosphate (S1P) to remove dying or surplus cells while maintaining the epithelium barrier. Both types start with the activation of S1P receptor type 2, located in neighboring cells. In this work, we studied the effect of cell differentiation induced by hypertonicity on cell extrusion in MDCK cells, and we provide new insights into the associated molecular mechanism. We found that the different stages of differentiation influence the rate of apoptotic cell extrusion. Besides, we used a novel methodology to demonstrate that S1P increase in extruding cells of differentiated monolayers. These results show for first time that cell extrusion is triggered by the single-cell synthesis of S1P by sphingosine kinase 2 (SphK2), but not SphK1, of the extruding cell itself. Moreover, the inhibition or knockdown of SphK2 prevents cell extrusion and cell-cell junction protein degradation, but not apoptotic nuclear fragmentation. Thus, we propose SphK2 as the biochemical key to ensure the preservation of the epithelial barrier under hypertonic stress.


Sujet(s)
Apoptose , Lysophospholipides/métabolisme , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Sphingosine/analogues et dérivés , Animaux , Différenciation cellulaire , Chiens , Rein/cytologie , Rein/métabolisme , Cellules rénales canines Madin-Darby , Analyse sur cellule unique , Sphingosine/métabolisme
15.
J Lipid Res ; 62: 100037, 2021.
Article de Anglais | MEDLINE | ID: mdl-32948663

RÉSUMÉ

Sphingolipids have emerged as bioactive lipids involved in the regulation of many physiological and pathological processes. In the retina, they have been established to participate in numerous processes, such as neuronal survival and death, proliferation and migration of neuronal and vascular cells, inflammation, and neovascularization. Dysregulation of sphingolipids is therefore crucial in the onset and progression of retinal diseases. This review examines the involvement of sphingolipids in retinal physiology and diseases. Ceramide (Cer) has emerged as a common mediator of inflammation and death of neuronal and retinal pigment epithelium cells in animal models of retinopathies such as glaucoma, age-related macular degeneration (AMD), and retinitis pigmentosa. Sphingosine-1-phosphate (S1P) has opposite roles, preventing photoreceptor and ganglion cell degeneration but also promoting inflammation, fibrosis, and neovascularization in AMD, glaucoma, and pro-fibrotic disorders. Alterations in Cer, S1P, and ceramide 1-phosphate may also contribute to uveitis. Notably, use of inhibitors that either prevent Cer increase or modulate S1P signaling, such as Myriocin, desipramine, and Fingolimod (FTY720), preserves neuronal viability and retinal function. These findings underscore the relevance of alterations in the sphingolipid metabolic network in the etiology of multiple retinopathies and highlight the potential of modulating their metabolism for the design of novel therapeutic approaches.


Sujet(s)
Sphingolipides
16.
Exp Eye Res ; 202: 108359, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-33197453

RÉSUMÉ

Müller glial cells, the major glial cell type in the retina, are activated by most retina injuries, leading to an increased proliferation and migration that contributes to visual dysfunction. The molecular cues involved in these processes are still ill defined. We demonstrated that sphingosine-1-phosphate (S1P), a bioactive sphingolipid, promotes glial migration. We now investigated whether ceramide-1-phosphate (C1P), also a bioactive sphingolipid, was involved in Müller glial cell migration. We evaluated cell migration in primary Müller glial cultures, prepared from newborn rat retinas, by the scratch wound assay. Addition of either 10 µM C8-ceramide-1-phosphate (C8-C1P) or 5 µM C16-C1P (a long chain, natural C1P) stimulated glial migration. Inhibiting PI3K almost completely blocked C8-C1P-elicited migration whereas inhibition of ERK1-2/MAPK pathway diminished it and p38MAPK inhibition did not affect it. Pre-treatment with a cytoplasmic phospholipase A2 (cPLA2) inhibitor markedly reduced C8-C1P-induced migration. Inhibiting ceramide kinase (CerK), the enzyme catalyzing C1P synthesis, partially decreased glial migration. Combined addition of S1P and C8-C1P promoted glial migration to the same extent as when they were added separately, suggesting they converge on their downstream signaling to stimulate Müller glia migration. These results suggest that C1P addition stimulated migration of glial Müller cells, promoting the activation of cPLA2, and the PI3K and ERK/MAPK pathways. They also suggest that CerK-dependent C1P synthesis was one of the factors contributing to glial migration, thus uncovering a novel role for C1P in controlling glial motility.


Sujet(s)
Céramides/pharmacologie , Cellules épendymogliales/cytologie , Cellules ganglionnaires rétiniennes/cytologie , Animaux , Animaux nouveau-nés , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cellules épendymogliales/effets des médicaments et des substances chimiques , Modèles animaux , Rats , Rat Wistar , Cellules ganglionnaires rétiniennes/effets des médicaments et des substances chimiques , Transduction du signal
17.
Front Pharmacol ; 11: 628, 2020.
Article de Anglais | MEDLINE | ID: mdl-32457622

RÉSUMÉ

GPR12 is a G protein-coupled orphan receptor genetically related to type 1 and type 2 cannabinoid receptors (CB1 and CB2) which are ancient proteins expressed all over the body. Both cannabinoid receptors, but especially CB1, are involved in neurodevelopment and cognitive processes such as learning, memory, brain reward, coordination, etc. GPR12 shares with CB1 that both are mainly expressed into the brain. Regrettably, very little is known about physiology of GPR12. Concerning its pharmacology, GPR12 seems to be endogenously activated by the lysophospholipids sphingosine-1-phosphate (S1P) and sphingosyl-phosphorylcholine (SPC). Exogenously, GPR12 is a target for the phytocannabinoid cannabidiol (CBD). Functionally, GPR12 seems to be related to neurogenesis and neural inflammation, but its relationship with cognitive functions remains to be characterized. Although GPR12 was initially suggested to be a cannabinoid receptor, it does not meet the five criteria proposed in 2010 by the International Union of Basic and Clinical Pharmacology (IUPHAR). In this review, we analyze all the direct available information in PubMed database about expression, function, and pharmacology of this receptor in central nervous system (CNS) trying to provide a broad overview of its current and prospective neurophysiology. Moreover, in this mini-review we highlight the need to produce more relevant data about the functions of GPR12 in CNS. Hence, this work should motivate further research in this field.

18.
Cells ; 9(3)2020 03 01.
Article de Anglais | MEDLINE | ID: mdl-32121501

RÉSUMÉ

For decades, sphingolipids have been related to several biological functions such as immune system regulation, cell survival, and proliferation. Recently, it has been reported that sphingolipids could be biomarkers in cancer and in other human disorders such as metabolic diseases. This is evidenced by the biological complexity of the sphingolipids associated with cell type-specific signaling and diverse sphingolipids molecules. As mitochondria dynamics have serious implications in homeostasis, in the present review, we focused on the relationship between sphingolipids, mainly ceramides and sphingosine-1-phosphate, and mitochondrial dynamics directed by fission, fusion, and mitophagy. There is evidence that the balances of ceramides (C18 and C16) and S1P, as well as the location of specific ceramide synthases in mitochondria, have roles in mitophagy and fission with an impact on cell fate and metabolism. However, signaling pathways controlling the sphingolipids metabolism and their location in mitochondria need to be better understood in order to propose new interventions and therapeutic strategies.


Sujet(s)
Dynamique mitochondriale/immunologie , Transduction du signal/immunologie , Sphingolipides/métabolisme , Humains
19.
Expert Opin Biol Ther ; 20(4): 413-420, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32093531

RÉSUMÉ

Introduction: Sphingosine-1-phosphate (S1P) is a membrane-derived lysophospholipid signaling molecule implicated in various physiological and pathological processes, such as regulation of the immune, cardiovascular, pulmonary, and nervous systems and theoretical cancer-related risks, through extracellular activation of S1P1-5 receptors.Areas covered: S1P receptor agonism is a novel strategy for the treatment of UC targeting lymphocyte recirculation, through blockade of lymphocyte egress from lymph nodes. We conducted an extensive literature review on PUBMED on currently available data on molecular aspects of S1P modulation, the mechanisms of action of S1PR agonists (fingolimod, ozanimod, etrasimod, and KRP-203), and their potential efficacy and safety for the treatment of patients with ulcerative colitis.Expert opinion: Selective S1P modulators have emerged to enlarge the efficacy and safety profile of this class of agents. Phase 3 programs should add the potential body of evidence to prove their benefit for the management of UC patients.


Sujet(s)
Rectocolite hémorragique/traitement médicamenteux , Modulateurs des récepteurs de la sphingosine 1 phosphate/usage thérapeutique , Récepteurs de la sphingosine-1-phosphate/agonistes , Acétates/usage thérapeutique , Essais cliniques comme sujet , Rectocolite hémorragique/anatomopathologie , Humains , Indanes/usage thérapeutique , Indoles/usage thérapeutique , Lysophospholipides/métabolisme , Oxadiazoles/usage thérapeutique , Sphingosine/analogues et dérivés , Sphingosine/métabolisme , Récepteurs de la sphingosine-1-phosphate/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/métabolisme
20.
Bioorg Chem ; 94: 103414, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31757412

RÉSUMÉ

Sphingosine-1-phosphate is now emerging as an important player in cancer, inflammation, autoimmune, neurological and cardiovascular disorders. Abundance evidence in animal and humans cancer models has shown that SphK1 is linked to cancer. Thus, there is a great interest in the development new SphK1 inhibitors as a potential new treatment for cancer. In a search for new SphK1 inhibitors we selected the well-known SKI-II inhibitor as the starting structure and we synthesized a new inhibitor structurally related to SKI-II with a significant but moderate inhibitory effect. In a second approach, based on our molecular modeling results, we designed new structures based on the structure of PF-543, the most potent known SphK1 inhibitor. Using this approach, we report the design, synthesis and biological evaluation of a new series of compounds with inhibitory activity against both SphK1 and SphK2. These new inhibitors were obtained incorporating new connecting chains between their polar heads and hydrophobic tails. On the other hand, the combined techniques of molecular dynamics simulations and QTAIM calculations provided complete and detailed information about the molecular interactions that stabilize the different complexes of these new inhibitors with the active sites of the SphK1. This information will be useful in the design of new SphK inhibitors.


Sujet(s)
Phosphotransferases (Alcohol Group Acceptor)/antagonistes et inhibiteurs , Pyrimidines/usage thérapeutique , Conception de médicament , Humains , Modèles moléculaires , Pyrimidines/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE