Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Phytomedicine ; 128: 155390, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38569296

RESUMEN

BACKGROUND: Well-defined and effective pharmacological interventions for clinical management of myocardial ischemia/reperfusion (MI/R) injury are currently unavailable. Shexiang Baoxin Pill (SBP), a traditional Chinese medicine Previous research on SBP has been confined to single-target treatments for MI/R injury, lacking a comprehensive examination of various aspects of MI/R injury and a thorough exploration of its underlying mechanisms. PURPOSE: This study aimed to investigate the therapeutic potential of SBP for MI/R injury and its preventive effects on consequent chronic heart failure (CHF). Furthermore, we elucidated the specific mechanisms involved, contributing valuable insights into the potential pharmacological interventions for the clinical treatment of MI/R injury. METHODS: We conducted a comprehensive identification of SBP components using high-performance liquid chromatography. Subsequently, we performed a network pharmacology analysis based on the identification results, elucidating the key genes influenced by SBP. Thereafter, through bioinformatics analysis of the key genes and validation through mRNA and protein assays, we ultimately determined the centralized upstream targets. Lastly, we conducted in vitro experiments using myocardial and endothelial cells to elucidate and validate potential underlying mechanisms. RESULTS: SBP can effectively mitigate cell apoptosis, oxidative stress, and inflammation, as well as promote vascular regeneration following MI/R, resulting in improved cardiac function and reduced CHF risk. Mechanistically, SBP treatment upregulates sphingosine-1-phosphate receptor 1 (S1PR1) expression and activates the S1PR1 signaling pathway, thereby regulating the expression of key molecules, including phosphorylated Protein Kinase B (AKT), phosphorylated signal transducer and activator of transcription 3, epidermal growth factor receptor, vascular endothelial growth factor A, tumor necrosis factor-α, and p53. CONCLUSION: This study elucidated the protective role of SBP in MI/R injury and its potential to reduce the risk of CHF. Furthermore, by integrating downstream effector proteins affected by SBP, this research identified the upstream effector protein S1PR1, enhancing our understanding of the pharmacological characteristics and mechanisms of action of SBP. The significance of this study lies in providing compelling evidence for the use of SBP as a traditional Chinese medicine for MI/R injury and consequent CHF prevention.


Asunto(s)
Medicamentos Herbarios Chinos , Insuficiencia Cardíaca , Daño por Reperfusión Miocárdica , Receptores de Esfingosina-1-Fosfato , Animales , Humanos , Masculino , Ratones , Apoptosis/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Insuficiencia Cardíaca/tratamiento farmacológico , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Farmacología en Red , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Receptores de Esfingosina-1-Fosfato/metabolismo
2.
Front Immunol ; 13: 801727, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35720286

RESUMEN

Background: Results from our recent study demonstrate that sphingosine-1-phosphate receptor 1 (S1PR1) modulation improves microvascular hemodynamics after cerebrovascular thrombosis. This study was to determine the microvascular hemodynamic effects of a sub-thrombolytic dose of tPA combined with a selective S1PR1 modulator ozanimod in a mouse model of cerebrovascular thrombosis. Methods: Microvascular circulation in mice was monitored in vivo by two-photon microscopy. Thrombosis was induced in cortical arterioles by laser irradiation. Arteriolar flow velocity was measured by line-scanning following plasma-labeling with fluorescein-dextran. Results: Laser-induced thrombosis led to a persistent reduction of flow velocity in cortical arterioles. Sub-thrombolytic dose of tPA along with vehicle control did not improve the flow velocity in cortical arterioles following thrombosis. In contrast, a sub-thrombolytic dose of tPA along with ozanimod dramatically restored flow velocity in cortical arterioles following thrombosis. Ozanimod did not affect coagulation and bleeding time. Notably, ozanimod reduced thrombus volume without altering microvascular lumen diameter. In addition, ozanimod reduced leukocyte components within the thrombus. Conclusions: These findings demonstrate that the thrombolytic effect of a sub-thrombolytic dose of tPA is markedly enhanced by S1PR1 modulation, implying that S1PR1 modulation may improve the therapeutic benefit of low-dose tPA in patients with acute ischemic stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico , Trombosis , Animales , Fibrinolíticos/uso terapéutico , Humanos , Ratones , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Terapia Trombolítica , Trombosis/tratamiento farmacológico , Activador de Tejido Plasminógeno/farmacología , Activador de Tejido Plasminógeno/uso terapéutico
3.
Int J Mol Sci ; 22(24)2021 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-34948163

RESUMEN

Although numerous experiments revealed an essential role of a lipid mediator, sphingosine-1-phosphate (S1P), in breast cancer (BC) progression, the clinical significance of S1P remains unclear due to the difficulty of measuring lipids in patients. The aim of this study was to determine the plasma concentration of S1P in estrogen receptor (ER)-positive BC patients, as well as to investigate its clinical significance. We further explored the possibility of a treatment strategy targeting S1P in ER-positive BC patients by examining the effect of FTY720, a functional antagonist of S1P receptors, on hormone therapy-resistant cells. Plasma S1P levels were significantly higher in patients negative for progesterone receptor (PgR) expression than in those positive for expression (p = 0.003). Plasma S1P levels were also significantly higher in patients with larger tumor size (p = 0.012), lymph node metastasis (p = 0.014), and advanced cancer stage (p = 0.003), suggesting that higher levels of plasma S1P are associated with cancer progression. FTY720 suppressed the viability of not only wildtype MCF-7 cells, but also hormone therapy-resistant MCF-7 cells. Targeting S1P signaling in ER-positive BC appears to be a possible new treatment strategy, even for hormone therapy-resistant patients.


Asunto(s)
Neoplasias de la Mama/metabolismo , Lisofosfolípidos/análisis , Esfingosina/análogos & derivados , Adulto , Anciano , Biomarcadores de Tumor/sangre , Neoplasias de la Mama/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Clorhidrato de Fingolimod/farmacología , Expresión Génica/genética , Humanos , Metástasis Linfática , Lisofosfolípidos/sangre , Lisofosfolípidos/metabolismo , Células MCF-7 , Persona de Mediana Edad , Plasma/química , Receptores de Estrógenos/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Esfingosina/análisis , Esfingosina/sangre , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Receptores de Esfingosina-1-Fosfato/metabolismo
4.
Biomed Pharmacother ; 143: 112197, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34560541

RESUMEN

The sphingolipids ceramide (Cer), ceramide-1-phosphate (C1P), sphingosine (Sph), and sphingosine-1-phosphate (S1P)) are key signaling molecules that regulate many patho-biological processes. During the last decade, they have gained increasing attention since they may participate in important and numerous retinal processes, such as neuronal survival and death, proliferation and migration of neuronal and vascular cells, inflammation, and neovascularization. Cer for instance has emerged as a key mediator of inflammation and death of neuronal and retinal pigment epithelium cells in experimental models of retinopathies such as glaucoma, age-related macular degeneration (AMD), and retinitis pigmentosa. S1P may have opposite biological actions, 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. Furthermore, use of inhibitors that either prevent Cer increase or modulate S1P signaling, such as Myriocin, desipramine, and Fingolimod (FTY720), have been shown to preserve neuronal viability and retinal function. Collectively, the expanding role for these sphingolipids in the modulation of vital processes in retina cell types and in their dysregulation in retinal degenerations makes them attractive therapeutic targets.


Asunto(s)
Retina/metabolismo , Enfermedades de la Retina/metabolismo , Esfingolípidos/metabolismo , Animales , Ceramidas/metabolismo , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Lisofosfolípidos/metabolismo , Terapia Molecular Dirigida , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/efectos de los fármacos , Retina/patología , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Moduladores de los Receptores de fosfatos y esfingosina 1/uso terapéutico , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Receptores de Esfingosina-1-Fosfato/metabolismo
5.
Sci Rep ; 11(1): 15308, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321503

RESUMEN

Sphingosine 1-phosphate (S1P) is a bioactive signalling sphingolipid that is increased in diseases such as obesity and diabetes. S1P can modulate platelet function, however the direction of effect and S1P receptors (S1PRs) involved are controversial. Here we describe the role of S1P in regulating human platelet function and identify the receptor subtypes responsible for S1P priming. Human platelets were treated with protease-activated receptor 1 (PAR-1)-activating peptide in the presence or absence of S1P, S1PR agonists or antagonists, and sphingosine kinases inhibitors. S1P alone did not induce platelet aggregation but at low concentrations S1P enhanced PAR1-mediated platelet responses, whereas PAR1 responses were inhibited by high concentrations of S1P. This biphasic effect was mimicked by pan-S1PR agonists. Specific agonists revealed that S1PR1 receptor activation has a positive priming effect, S1PR2 and S1PR3 have no effect on platelet function, whereas S1PR4 and S1PR5 receptor activation have an inhibitory effect on PAR-1 mediated platelet function. Although platelets express both sphingosine kinase 1/2, enzymes which phosphorylate sphingosine to produce S1P, only dual and SphK2 inhibition reduced platelet function. These results support a role for SphK2-mediated S1P generation in concentration-dependent positive and negative priming of platelet function, through S1PR1 and S1PR4/5 receptors, respectively.


Asunto(s)
Lisofosfolípidos/farmacología , Activación Plaquetaria/efectos de los fármacos , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Esfingosina/análogos & derivados , Plaquetas/efectos de los fármacos , Plaquetas/ultraestructura , Proteínas Portadoras/farmacología , Forma de la Célula/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Lisofosfolípidos/agonistas , Lisofosfolípidos/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Agregación Plaquetaria/efectos de los fármacos , Receptor PAR-1/agonistas , Esfingosina/agonistas , Esfingosina/antagonistas & inhibidores , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato/fisiología
6.
J Clin Pharmacol ; 61(1): 82-89, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32656870

RESUMEN

Half-life is a standard result reported with analysis of pharmacokinetic data. Different definitions such as noncompartmental half-life, terminal half-life, effective half-life, and context-sensitive half-life can yield substantially different estimates of the quantity "half-life." Time to attainment of steady-state conditions is generally derived from (terminal) half-life and therefore sensitive toward the definition of half-life. Thus, estimates of the time to attain steady state must be provided with a precise definition of steady state and the method used for estimation, particularly for drugs with long (terminal) half-life. For clinical purposes, terminal half-life can have limited relevance if drug concentrations in the terminal elimination phase are low. A general rule for which half-life to use is infeasible. While limited accumulation can be negligible if a plateau in pharmacokinetics/pharmacodynamics is reached or with a wide therapeutic window (ie, exposure range), small additional drug accumulation can be highly relevant for drugs with a narrow therapeutic window. Beyond the average, estimation of individual time to attainment of steady state can add highly relevant information about the variability between subjects. Simulations from population models and the use of different definitions of steady state provide an assessment of robustness of the results. The relevance of accurate estimation of time to attainment of steady state is illustrated with cenerimod, an sphingosine-1-phosphate 1 receptor modulator with long half-life currently in clinical development for which estimates of time to steady state ranged from 35 to 110 days with different calculations.


Asunto(s)
Semivida , Farmacocinética , Humanos , Oxadiazoles/farmacocinética , Glicoles de Propileno/farmacocinética , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Factores de Tiempo
7.
Neuromolecular Med ; 23(1): 47-67, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33180310

RESUMEN

Sphingosine 1-phosphates (S1Ps) are bioactive lipids that mediate a diverse range of effects through the activation of cognate receptors, S1P1-S1P5. Scrutiny of S1P-regulated pathways over the past three decades has identified important and occasionally counteracting functions in the brain and cerebrovascular system. For example, while S1P1 and S1P3 mediate proinflammatory effects on glial cells and directly promote endothelial cell barrier integrity, S1P2 is anti-inflammatory but disrupts barrier integrity. Cumulatively, there is significant preclinical evidence implicating critical roles for this pathway in regulating processes that drive cerebrovascular disease and vascular dementia, both being part of the continuum of vascular cognitive impairment (VCI). This is supported by clinical studies that have identified correlations between alterations of S1P and cognitive deficits. We review studies which proposed and evaluated potential mechanisms by which such alterations contribute to pathological S1P signaling that leads to VCI-associated chronic neuroinflammation and neurodegeneration. Notably, S1P receptors have divergent but overlapping expression patterns and demonstrate complex interactions. Therefore, the net effect produced by S1P represents the cumulative contributions of S1P receptors acting additively, synergistically, or antagonistically on the neural, vascular, and immune cells of the brain. Ultimately, an optimized therapeutic strategy that targets S1P signaling will have to consider these complex interactions.


Asunto(s)
Demencia Vascular/fisiopatología , Lisofosfolípidos/fisiología , Receptores de Esfingosina-1-Fosfato/fisiología , Esfingosina/análogos & derivados , Aldehído-Liasas/antagonistas & inhibidores , Aldehído-Liasas/fisiología , Enfermedad de Alzheimer/fisiopatología , Animales , Trastornos Cerebrovasculares/fisiopatología , Ensayos Clínicos como Asunto , Sistemas de Liberación de Medicamentos , Evaluación Preclínica de Medicamentos , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/fisiopatología , Inflamación , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/fisiopatología , Ratones , Ratones Noqueados , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/fisiopatología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Transducción de Señal , Esfingosina/fisiología , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos
8.
Nat Chem Biol ; 17(2): 169-177, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32929277

RESUMEN

Many intracellular proteins are modified by N-acetylglucosamine, a post-translational modification termed O-GlcNAc. This modification is found on serine and threonine side chains and has the potential to regulate signaling pathways through interplay with phosphorylation. Here, we discover and characterize one such example. We find that O-GlcNAc levels control the sensitivity of fibroblasts to actin contraction induced by the signaling lipid sphingosine-1-phosphate (S1P), culminating in the phosphorylation of myosin light chain (MLC) and cellular contraction. Specifically, O-GlcNAc modification of the phosphatase subunit MYPT1 inhibits this pathway by blocking MYPT1 phosphorylation, maintaining its activity and causing the dephosphorylation of MLC. Finally, we demonstrate that O-GlcNAc levels alter the sensitivity of primary human dermal fibroblasts in a collagen-matrix model of wound healing. Our findings have important implications for the role of O-GlcNAc in fibroblast motility and differentiation, particularly in diabetic wound healing.


Asunto(s)
Acetilglucosamina/genética , Lisofosfolípidos/farmacología , Fosfatasa de Miosina de Cadena Ligera/genética , Esfingosina/análogos & derivados , Actinas/fisiología , Animales , Citoesqueleto/efectos de los fármacos , Fibroblastos , Técnicas de Silenciamiento del Gen , Glucosa/farmacología , Ratones , Contracción Muscular/efectos de los fármacos , Células 3T3 NIH , Fosforilación , Procesamiento Proteico-Postraduccional , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato/agonistas , Receptores de Esfingosina-1-Fosfato/antagonistas & inhibidores , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos
10.
Int J Mol Sci ; 21(18)2020 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-32899717

RESUMEN

Multiple sclerosis (MS) is a chronic, inflammatory, autoimmune disease of the central nervous system (CNS) which is associated with lower life expectancy and disability. The experimental antigen-induced encephalomyelitis (EAE) in mice is a useful animal model of MS, which allows exploring the etiopathogenetic mechanisms and testing novel potential therapeutic drugs. A new therapeutic paradigm for the treatment of MS was introduced in 2010 through the sphingosine 1-phosphate (S1P) analogue fingolimod (FTY720, Gilenya®), which acts as a functional S1P1 antagonist on T lymphocytes to deplete these cells from the blood. In this study, we synthesized two novel structures, ST-1893 and ST-1894, which are derived from fingolimod and chemically feature a morpholine ring in the polar head group. These compounds showed a selective S1P1 activation profile and a sustained S1P1 internalization in cultures of S1P1-overexpressing Chinese hamster ovary (CHO)-K1 cells, consistent with a functional antagonism. In vivo, both compounds induced a profound lymphopenia in mice. Finally, these substances showed efficacy in the EAE model, where they reduced clinical symptoms of the disease, and, on the molecular level, they reduced the T-cell infiltration and several inflammatory mediators in the brain and spinal cord. In summary, these data suggest that S1P1-selective compounds may have an advantage over fingolimod and siponimod, not only in MS but also in other autoimmune diseases.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Clorhidrato de Fingolimod/farmacología , Morfolinos/farmacología , Animales , Células CHO , Sistema Nervioso Central/efectos de los fármacos , Cricetulus , Modelos Animales de Enfermedad , Encefalomielitis/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Clorhidrato de Fingolimod/análogos & derivados , Inmunosupresores/uso terapéutico , Ligandos , Linfopenia/tratamiento farmacológico , Lisofosfolípidos/metabolismo , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Receptores de Esfingosina-1-Fosfato/metabolismo , Médula Espinal/efectos de los fármacos , Linfocitos T/efectos de los fármacos
11.
Curr Opin Support Palliat Care ; 14(2): 107-111, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32349095

RESUMEN

PURPOSE OF REVIEW: The treatment of cancer-induced bone pain (CIBP) has been proven ineffective and relies heavily on opioids, the target of highly visible criticism for their negative side effects. Alternative therapeutic agents are needed and the last few years have brought promising results, detailed in this review. RECENT FINDINGS: Cysteine/glutamate antiporter system, xc, cannabinoids, kappa opioids, and a ceramide axis have all been shown to have potential as novel therapeutic targets without the negative effects of opioids. SUMMARY: Review of the most recent and promising studies involving CIBP, specifically within murine models. Cancer pain has been reported by 30-50% of all cancer patients and even more in late stages, however the standard of care is not effective to treat CIBP. The complicated and chronic nature of this type of pain response renders over the counter analgesics and opioids largely ineffective as well as difficult to use due to unwanted side effects. Preclinical studies have been standardized and replicated while novel treatments have been explored utilizing various alternative receptor pathways: cysteine/glutamate antiporter system, xc, cannabinoid type 1 receptor, kappa opioids, and a ceramide axis sphingosine-1-phosphate/sphingosine-1-phosphate receptor 1.


Asunto(s)
Huesos/fisiopatología , Dolor en Cáncer/tratamiento farmacológico , Animales , Antiportadores/efectos de los fármacos , Antiportadores/metabolismo , Huesos/inervación , Cannabinoides/uso terapéutico , Modelos Animales de Enfermedad , Humanos , Ratones , Receptor Cannabinoide CB1/efectos de los fármacos , Receptor Cannabinoide CB1/metabolismo , Receptores Opioides kappa/efectos de los fármacos , Receptores Opioides kappa/metabolismo , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Receptores de Esfingosina-1-Fosfato/metabolismo
12.
Life Sci ; 249: 117542, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32169519

RESUMEN

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that regulates lymphocyte trafficking, glial cell activation, vasoconstriction, endothelial barrier function, and neuronal death pathways in the brain. Research has increasingly implicated S1P in the pathology of cerebral ischemia reperfusion (IR) injury. As a high-affinity agonist of S1P receptor, fingolimod exhibits excellent neuroprotective effects against ischemic challenge both in vivo and in vitro. By summarizing recent progress on how S1P participates in the development of brain IR injury, this review identifies potential therapeutic targets for the treatment of brain IR injury.


Asunto(s)
Encéfalo/irrigación sanguínea , Clorhidrato de Fingolimod/uso terapéutico , Lisofosfolípidos/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Esfingosina/análogos & derivados , Animales , Humanos , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo
13.
Lancet Gastroenterol Hepatol ; 5(9): 850-861, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32171056

RESUMEN

The incidence and prevalence of ulcerative colitis are increasing globally. Although the exact cause and pathogenesis of this disease is unclear, research has led to a better understanding of the condition and to identification of new targets for therapy, which in turn has encouraged the development of new therapies. As well as biologic therapies, which have changed the way inflammatory bowel disease is managed, small molecules have been developed for the treatment of ulcerative colitis. These small molecule treatments are orally administered and are likely to bring a substantial shift in the way this chronic disease is treated. Oral therapies offer many advantages over infusion therapies, such as ease of use, increased acceptability by patients, and reduction of cost. This Review focuses not only on oral therapies that have been approved for use in ulcerative colitis, but also on those that are in development, providing a comprehensive overview for clinicians of available oral therapies and drugs that are likely to become available. We have also reviewed drugs that have shown promise in preclinical studies and could be effective future therapies.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Terapia Molecular Dirigida/métodos , Administración Oral , Anciano , Enfermedad Crónica/tratamiento farmacológico , Ensayos Clínicos como Asunto , Colitis Ulcerosa/epidemiología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Inmunomodulación , Incidencia , Inhibidores de las Cinasas Janus/farmacología , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/efectos de los fármacos , Persona de Mediana Edad , Inhibidores de Fosfodiesterasa 4/farmacología , Prevalencia , Receptores de Esfingosina-1-Fosfato/agonistas , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos
14.
Circ Heart Fail ; 12(11): e005835, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31684756

RESUMEN

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is a genetic cardiovascular disorder, primarily involving mutations in sarcomeric proteins. HCM patients present with hypertrophy, diastolic dysfunction, and fibrosis, but there is no specific treatment. The sphingosine-1-phosphate receptor modulator, FTY720/fingolimod, is approved for treatment of multiple sclerosis. We hypothesize that modulation of the sphingosine-1-phosphate receptor by FTY720 would be of therapeutic benefit in sarcomere-linked HCM. METHODS: We treated mice with an HCM-linked mutation in tropomyosin (Tm-E180G) and nontransgenic littermates with FTY720 or vehicle for 6 weeks. Compared with vehicle-treated, FTY720-treated Tm-E180G mice had a significant reduction in left atrial size (1.99±0.19 [n=7] versus 2.70±0.44 [n=6] mm; P<0.001) and improvement in diastolic function (E/A ratio: 2.69±0.38 [n=7] versus 5.34±1.19 [n=6]; P=0.004) as assessed by echocardiography. RESULTS: Pressure-volume relations revealed significant improvements in the end-diastolic pressure volume relationship, relaxation kinetics, preload recruitable stroke work, and ejection fraction. Detergent-extracted fiber bundles revealed a significant decrease in myofilament Ca2+-responsiveness (pCa50=6.15±0.11 [n=13] versus 6.24±0.06 [n=14]; P=0.041). We attributed these improvements to a downregulation of S-glutathionylation of cardiac myosin binding protein-C in FTY720-treated Tm-E180G mice and reduction in oxidative stress by downregulation of NADPH oxidases with no changes in fibrosis. CONCLUSIONS: This is the first demonstration that modulation of S1PR results in decreased myofilament-Ca2+-responsiveness and improved diastolic function in HCM. We associated these changes with decreased oxidative modification of myofilament proteins via downregulation of NOX2. Our data support the hypothesis that modification of sphingolipid signaling may be a novel therapeutic approach in HCM.


Asunto(s)
Función del Atrio Izquierdo/efectos de los fármacos , Remodelación Atrial/efectos de los fármacos , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Clorhidrato de Fingolimod/farmacología , Miocitos Cardíacos/efectos de los fármacos , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Animales , Señalización del Calcio/efectos de los fármacos , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/fisiopatología , Diástole , Modelos Animales de Enfermedad , Femenino , Fibrosis , Predisposición Genética a la Enfermedad , Masculino , Ratones Mutantes , Mutación , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miofibrillas/efectos de los fármacos , Miofibrillas/metabolismo , Miofibrillas/patología , Estrés Oxidativo/efectos de los fármacos , Fenotipo , Receptores de Esfingosina-1-Fosfato/metabolismo , Tropomiosina/genética
15.
Expert Opin Investig Drugs ; 28(12): 1051-1057, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31603362

RESUMEN

Introduction: Multiple sclerosis (MS) causes focal lesions of immune-mediated demyelinating events followed by slow progressive accumulation of disability. Over the past 2 decades, multiple medications have been studied and approved for use in MS. Most of these agents work by modulating or suppressing the peripheral immune system. Siponimod is a newer-generation sphingosine 1 phosphate (S1P) receptor modulator that internalizes S1P1 receptors, thereby inhibiting efflux of lymphocytes from lymph nodes and thymus. There are promising data suggesting that it may also have a direct neuroprotective property independent of peripheral lymphocytopenia.Areas covered: We reviewed the pharmacology and the clinical and radiological effects of siponimod.Expert opinion: The selective effect of siponimod on the S1P1 and S1P5 receptors offers a favorable side-effect profile and transient bradycardia can be avoided by dose titration. A phase-II study showed that siponomod has dose-dependent beneficial effects in patients with relapsing remitting disease. The results of a phase-III study suggest that siponimod may be beneficial in secondary progressive MS, at least in patients with disease activity.


Asunto(s)
Azetidinas/administración & dosificación , Compuestos de Bencilo/administración & dosificación , Esclerosis Múltiple/tratamiento farmacológico , Moduladores de los Receptores de fosfatos y esfingosina 1/administración & dosificación , Animales , Azetidinas/efectos adversos , Azetidinas/farmacología , Compuestos de Bencilo/efectos adversos , Compuestos de Bencilo/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Esclerosis Múltiple/fisiopatología , Moduladores de los Receptores de fosfatos y esfingosina 1/efectos adversos , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Receptores de Esfingosina-1-Fosfato/metabolismo
16.
J Cell Physiol ; 234(10): 18029-18040, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30843214

RESUMEN

Fingolimod (FTY720) after phosphorylation, as the ligand of sphingosine 1-phosphate receptors (S1PRs), plays an important role in cell proliferation and differentiation. In this article, FTY720 in the treatment of coxsackievirus B3 (CVB3)-induced viral myocarditis was closely related to apoptosis and AKT/caspase-3 apoptotic pathways. We found that CVB3 inhibited myocardial apoptosis at the early stage with upregulating p-AKT level and downregulating activated caspase-3 level for replication of virus progeny, whereas it promoted apoptosis at a late stage with downregulating p-AKT and upregulating activated caspase-3 for releasing the newly synthesized virus to spread. Interestingly, FTY720 could reverse this trend; it promoted apoptosis at an early stage and inhibited apoptosis at the late stage in vivo and vitro, which proved the antiviral effect. We also found that S1PR1, S1PR4, and S1PR5, rather than S1PR2 and S1PR3, were regulated by FTY720 in this process. The results confirmed that FTY720 alleviates CVB3-induced myocarditis and inhibits viral replication through regulating S1PRs and AKT/caspase-3 pathways with a bidirectional regulation of apoptosis.


Asunto(s)
Antivirales/farmacología , Caspasa 3/metabolismo , Infecciones por Coxsackievirus/prevención & control , Enterovirus Humano B/efectos de los fármacos , Clorhidrato de Fingolimod/farmacología , Miocarditis/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/patología , Infecciones por Coxsackievirus/virología , Modelos Animales de Enfermedad , Enterovirus Humano B/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Masculino , Ratones Endogámicos BALB C , Miocarditis/metabolismo , Miocarditis/patología , Miocarditis/virología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Miocitos Cardíacos/virología , Transducción de Señal , Receptores de Esfingosina-1-Fosfato/metabolismo , Factores de Tiempo
17.
Brain Res Bull ; 146: 253-269, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30660717

RESUMEN

Hepatic encephalopathy (HE) is a complex neuropsychiatric disorder, which is associated with memory loss and behavioral abnormalities. The cellular and molecular mechanisms that led to hippocampal dysfunction in bile duct ligation (BDL)-induced HE and neuroprotective mechanisms of FTY720 administration were evaluated using whole-cell patch clamp, field-potential recording, western blot, stereology and behavioral experiments. The animals were divided into 4 groups of control (n = 24), sham (n = 21), BDL + V (n = 21) and BDL + FTY720 (n = 20), each having three subgroups. The first subgroup was used for field potential, western blot and stereology experiments. The second and third subgroups were used for behavioral experiments and whole cell patch clamp recording, respectively. The BDL led to considerable loss of hippocampal neurons and apoptosis, along with large impairments in their intrinsic electrophysiological properties, including decrease of firing frequency and increases of first spike latency (FSL), AHP amplitude, irregularity of firing, and half-width, as well as impaired long-term synaptic plasticity and memory. Importantly, FTY720 decreased AHP amplitude probably by direct inhibition of Ca2+ channels and/or KCa2+ currents and improved the FSL and firing irregularity and frequency possibly by decreasing A-type K+ currents in the BDL + FTY720 group. FTY720 administration in the BDL rats also decreased the release probability, which was evident by the increased paired-pulse ratio, but the increased number of readily releasable pool (RRP) of neurotransmitter. Moreover, the AHP improvement and RRP increment most likely led to recovery of LTP and memory performance. In total, FTY720 ameliorated brain disorders in the BDL rats via its direct neuroprotective and/or indirect anti-inflammatory effects.


Asunto(s)
Encefalopatía Hepática/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Animales , Conductos Biliares/patología , Región CA1 Hipocampal/metabolismo , Cognición , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod/farmacología , Encefalopatía Hepática/fisiopatología , Ligadura , Masculino , Trastornos de la Memoria , Plasticidad Neuronal , Ratas , Ratas Sprague-Dawley , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos
18.
J Immunol Res ; 2019: 5821589, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31930150

RESUMEN

Amiselimod (MT-1303) is a novel and selective sphingosine 1-phosphate receptor-1 (S1P1) modulator with a more favorable cardiac safety profile than other S1P1 receptor modulators. In this study, we evaluated the effects of MT-1303 on the progression of lupus nephritis in two well-known murine systemic lupus erythematosus (SLE) models, MRL/lpr and NZBWF1 mice, compared with those of FK506. Daily oral doses of 0.1 and 0.3 mg/kg MT-1303 not only inhibited the development of lupus nephritis when administered before onset in MRL/lpr and NZBWF1 mice but also improved symptoms of lupus nephritis when administered after onset in MRL/lpr mice. Its efficacy in these models was more potent or comparable to that of FK506 (1 and 3 mg/kg). In histological analysis, treatment with MT-1303 inhibited infiltration of T cells into the kidneys, mesangial expansion, and glomerular sclerosis. MT-1303 treatment resulted in a marked reduction in T cells and B cells in the peripheral blood and significantly inhibited increases in the number of plasma cells in the spleen and T cells in the kidneys. In addition, administration of MT-1303 suppressed elevations in serum anti-dsDNA antibody levels in MRL/lpr mice, but not in NZBWF1 mice. Our findings show that MT-1303 exhibits marked therapeutic effects on lupus nephritis in two SLE models, likely by reducing the infiltration of autoreactive T cells into the kidneys. These results suggest that MT-1303 has the potential to be used as a therapeutic agent for patients suffering from SLE, including lupus nephritis.


Asunto(s)
Riñón/efectos de los fármacos , Nefritis Lúpica/tratamiento farmacológico , Propanolaminas/uso terapéutico , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Animales , Anticuerpos Antinucleares/sangre , Anticuerpos Antinucleares/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Riñón/citología , Riñón/inmunología , Riñón/patología , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Ganglios Linfáticos/citología , Ganglios Linfáticos/efectos de los fármacos , Masculino , Células Mesangiales/efectos de los fármacos , Ratones , Ratones Endogámicos MRL lpr , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/inmunología , Receptores de Esfingosina-1-Fosfato/metabolismo , Bazo/citología , Bazo/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...