Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 322(1): L149-L161, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35015568

RESUMEN

Disruption of the lung endothelial barrier is a hallmark of acute respiratory distress syndrome (ARDS), for which no effective pharmacologic treatments exist. Prior work has demonstrated that FTY720 S-phosphonate (Tys), an analog of sphingosine-1-phosphate (S1P) and FTY720, exhibits potent endothelial cell (EC) barrier protective properties. In this study, we investigated the in vitro and in vivo efficacy of Tys against methicillin-resistant Staphylococcus aureus (MRSA), a frequent bacterial cause of ARDS. Tys-protected human lung EC from barrier disruption induced by heat-killed MRSA (HK-MRSA) or staphylococcal α-toxin and attenuated MRSA-induced cytoskeletal changes associated with barrier disruption, including actin stress fiber formation and loss of peripheral VE-cadherin and cortactin. Tys-inhibited Rho and myosin light chain (MLC) activation after MRSA and blocked MRSA-induced NF-κB activation and release of the proinflammatory cytokines, IL-6 and IL-8. In vivo, intratracheal administration of live MRSA in mice caused significant vascular leakage and leukocyte infiltration into the alveolar space. Pre- or posttreatment with Tys attenuated MRSA-induced lung permeability and levels of alveolar neutrophils. Posttreatment with Tys significantly reduced levels of bronchoalveolar lavage (BAL) VCAM-1 and plasma IL-6 and KC induced by MRSA. Dynamic intravital imaging of mouse lungs demonstrated Tys attenuation of HK-MRSA-induced interstitial edema and neutrophil infiltration into lung tissue. Tys did not directly inhibit MRSA growth or viability in vitro. In conclusion, Tys inhibits lung EC barrier disruption and proinflammatory signaling induced by MRSA in vitro and attenuates acute lung injury induced by MRSA in vivo. These results support the potential utility of Tys as a novel ARDS therapeutic strategy.


Asunto(s)
Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/patología , Permeabilidad de la Membrana Celular , Células Endoteliales/microbiología , Clorhidrato de Fingolimod/análogos & derivados , Staphylococcus aureus Resistente a Meticilina/fisiología , Organofosfonatos/farmacología , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Citoprotección/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Activación Enzimática/efectos de los fármacos , Clorhidrato de Fingolimod/farmacología , Humanos , Inflamación/patología , Ratones , Cadenas Ligeras de Miosina/metabolismo , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo
2.
Neuropharmacology ; 186: 108464, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33460688

RESUMEN

The sphingosine 1-phosphate (S1P) receptor 1 (S1P1) has emerged as a therapeutic target for the treatment of multiple sclerosis (MS). Fingolimod (FTY720) is the first functional antagonist of S1P1 that has been approved for oral treatment of MS. Previously, we have developed novel butterfly derivatives of FTY720 that acted similar to FTY720 in reducing disease symptoms in a mouse model of experimental autoimmune encephalomyelitis (EAE). In this study, we have synthesized a piperidine derivative of the oxazolo-oxazole compounds, denoted ST-1505, and its ring-opened analogue ST-1478, and characterised their in-vitro and in-vivo functions. Notably, the 3-piperidinopropyloxy moiety resembles a structural motif of pitolisant, a drug with histamine H3R antagonistic/inverse agonist activity approved for the treatment of narcolepsy. Both novel compounds exerted H3R affinities, and in addition, ST-1505 was characterised as a dual S1P1+3 agonist, whereas ST-1478 was a dual S1P1+5 agonist. Both multitargeting compounds were also active in mice and reduced the lymphocyte numbers as well as diminished disease symptoms in the mouse model of MS. The effect of ST-1478 was dependent on SK-2 activity suggesting that it is a prodrug like FTY720, but with a more selective S1P receptor activation profile, whereas ST-1505 is a fully active drug even in the absence of SK-2. In summary, these data suggest that the well soluble piperidine derivatives ST-1505 and ST-1478 hold promise as novel drugs for the treatment of MS and other autoimmune or inflammatory diseases, and by their H3R antagonist potency, they might additionally improve cognitive impairment during disease.


Asunto(s)
Encefalomielitis Autoinmune Experimental/prevención & control , Clorhidrato de Fingolimod/administración & dosificación , Antagonistas de los Receptores Histamínicos H3/administración & dosificación , Esclerosis Múltiple/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Receptores de Esfingosina-1-Fosfato/agonistas , Animales , Células CHO , Cricetinae , Cricetulus , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Clorhidrato de Fingolimod/análogos & derivados , Clorhidrato de Fingolimod/química , Antagonistas de los Receptores Histamínicos H3/química , Antagonistas de los Receptores Histamínicos H3/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Esclerosis Múltiple/metabolismo , Fármacos Neuroprotectores/química , Estructura Secundaria de Proteína , Receptores de Esfingosina-1-Fosfato/metabolismo
3.
Arch Pharm Res ; 43(10): 1046-1055, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33111965

RESUMEN

Fingolimod has been evaluated for use as an anticancer agent. However, many steps are required to synthesize fingolimod because of its intricate structure. A fingolimod analogue, N-(4-(2-((4-methoxybenzyl)amino)ethyl)phenyl)heptanamide (MPH), also has anti-cancer effects and is easier to synthesize but is poorly soluble in water. To compensate for its poor water solubility, MPH-loaded polymeric micelles were prepared by thin film hydration method using various polymers and the physicochemical properties of the MPH-loaded micelles such as particle size, drug-loading (DL, %), and encapsulation efficiency (EE, %) were evaluated. A storage stability test was conducted to select the final formulation and the release profile of the MPH-loaded micelles was confirmed by in vitro release assay. MPH-loaded mPEG-b-PLA micelles were selected for further testing based on their stability and physicochemical properties; they were stable for stable for 14 days at 4 °C and 25 °C and for 7 days at 37 °C. They showed anti-cancer efficacy against both A549 and U87 cancer cells. Encapsulation of MPH in polymeric micelles did not decrease the in vitro cytotoxicity of MPH. The findings of this study lay the groundwork for future formulations that enable the effective and stable delivery of poorly water-soluble agents.


Asunto(s)
Antineoplásicos/farmacocinética , Portadores de Fármacos/química , Clorhidrato de Fingolimod/farmacocinética , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Línea Celular Tumoral , Liberación de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Estabilidad de Medicamentos , Clorhidrato de Fingolimod/administración & dosificación , Clorhidrato de Fingolimod/análogos & derivados , Clorhidrato de Fingolimod/química , Humanos , Micelas , Tamaño de la Partícula , Polímeros/química , Solubilidad , Agua/química
4.
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
5.
J Parkinsons Dis ; 10(1): 185-192, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31561385

RESUMEN

BACKGROUND: Parkinson's disease (PD) and multiple system atrophy (MSA) patients often suffer from gastrointestinal (GI) dysfunction and GI dysbiosis (microbial imbalance). GI dysfunction also occurs in mouse models of PD and MSA. OBJECTIVES: To assess gut dysfunction and dysbiosis in PD subjects as compared to controls, identify potential shared microbial taxa in humans and mouse models of PD and MSA, and to assess the effects of potential therapies on mouse GI microbiota. METHODS: In this human pilot study, GI function was assessed by fecal consistency/frequency measured using the Bristol Stool Form Scale and GI transit time assessed using Sitzmarks pills and abdominal radiology. Human and mouse microbiota were analyzed by extracting fecal genomic DNA followed by 16S rRNA sequencing. RESULTS: In our PD patients genera Akkermansia significantly increased while a trend toward increased Bifidobacterium and decreased Prevotella was observed. Families Bacteroidaceae and Lachnospiraceae and genera Prevotella and Bacteroides were detected in both humans and PD mice, suggesting potential shared biomarkers. In mice treated with the approved multiple sclerosis drug, FTY720, or with our FTY720-Mitoxy-derivative, we saw that FTY720 had little effect while FTY720-Mitoxy increased beneficial Ruminococcus and decreased Rickenellaceae family. CONCLUSION: Akkermansia and Prevotellaceae data reported by others were replicated in our human pilot study suggesting the use of those taxa as potential biomarkers for PD diagnosis. The effect of FTY720-Mitoxy on taxa Rikenellaceae and Ruminococcus and the relevance of S24-7 await further evaluation. It also remains to be determined if mouse microbiota have predictive power for human subjects.


Asunto(s)
Disbiosis/microbiología , Clorhidrato de Fingolimod/farmacología , Microbioma Gastrointestinal , Inmunosupresores/farmacología , Microbiota , Atrofia de Múltiples Sistemas/microbiología , Enfermedad de Parkinson/microbiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Estreñimiento/fisiopatología , Modelos Animales de Enfermedad , Femenino , Clorhidrato de Fingolimod/administración & dosificación , Clorhidrato de Fingolimod/análogos & derivados , Microbioma Gastrointestinal/efectos de los fármacos , Motilidad Gastrointestinal/fisiología , Humanos , Inmunosupresores/administración & dosificación , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Proyectos Piloto , ARN Ribosómico 16S
6.
Exp Neurol ; 325: 113120, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31751571

RESUMEN

Multiple system atrophy (MSA) is a fatal disorder with no effective treatment. MSA pathology is characterized by α-synuclein (aSyn) accumulation in oligodendrocytes, the myelinating glial cells of the central nervous system (CNS). aSyn accumulation in oligodendrocytes forms the pathognomonic glial cytoplasmic inclusions (GCIs) of MSA. MSA aSyn pathology is also associated with motor and autonomic dysfunction, including an impaired ability to sweat. MSA patients have abnormal CNS expression of glial-cell-line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF). Our prior studies using the parent compound FTY720, a food and drug administration (FDA) approved immunosuppressive for multiple sclerosis, reveal that FTY720 protects parkinsonian mice by increasing BDNF. Our FTY720-derivative, FTY720-Mitoxy, is known to increase expression of oligodendrocyte BDNF, GDNF, and nerve growth factor (NGF) but does not reduce levels of circulating lymphocytes as it is not phosphorylated so cannot modulate sphingosine 1 phosphate receptors (S1PRs). To preclinically assess FTY720-Mitoxy for MSA, we used mice expressing human aSyn in oligodendrocytes under a 2,' 3'-cyclic nucleotide 3'-phosphodiesterase (CNP) promoter. CNP-aSyn transgenic (Tg) mice develop motor dysfunction between 7 and 9 mo, and progressive GCI pathology. Using liquid chromatography-mass spectrometry (LC-MS/MS) and enzymatic assays, we confirmed that FTY720-Mitoxy was stable and active. Vehicle or FTY720-Mitoxy (1.1 mg/kg/day) was delivered to wild type (WT) or Tg littermates from 8.5-11.5 mo by osmotic pump. We behaviorally assessed their movement by rotarod and sweat production by starch­iodine test. Postmortem tissues were evaluated by qPCR for BDNF, GDNF, NGF and GDNF-receptor RET mRNA and for aSyn, BDNF, GDNF, and Iba1 protein by immunoblot. MicroRNAs (miRNAs) were also assessed by qPCR. FTY720-Mitoxy normalized movement, sweat function and soleus muscle mass in 11.5 mo Tg MSA mice. FTY720-Mitoxy also increased levels of brain GDNF and reduced brain miR-96-5p, a miRNA that acts to decrease GDNF expression. Moreover, FTY720-Mitoxy blocked aSyn pathology measured by sequential protein extraction and immunoblot, and microglial activation assessed by immunohistochemistry and immunoblot. In the 3-nitropropionic acid (3NP) toxin model of MSA, FTY720-Mitoxy protected movement and mitochondria in WT and CNP-aSyn Tg littermates. Our data confirm potent in vivo protection by FTY720-Mitoxy, supporting its further evaluation as a potential therapy for MSA and related synucleinopathies.


Asunto(s)
Clorhidrato de Fingolimod/análogos & derivados , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Atrofia de Múltiples Sistemas/patología , Fármacos Neuroprotectores/farmacología , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Clorhidrato de Fingolimod/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/efectos de los fármacos , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Transgénicos , MicroARNs/efectos de los fármacos , MicroARNs/metabolismo , Atrofia de Múltiples Sistemas/metabolismo , Proteínas Proto-Oncogénicas c-ret/biosíntesis , Proteínas Proto-Oncogénicas c-ret/efectos de los fármacos , alfa-Sinucleína/genética
7.
Cancer Lett ; 468: 1-13, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31593801

RESUMEN

Acute myeloid leukemia (AML) is an aggressive disease associated with very poor prognosis. Most patients are older than 60 years, and in this group only 5-15% of cases survive over 5 years. Therefore, it is urgent to develop more effective targeted therapies. Inactivation of protein phosphatase 2 A (PP2A) is a recurrent event in AML, and overexpression of its endogenous inhibitor SET is detected in ~30% of patients. The PP2A activating drug FTY720 has potent anti-leukemic effects; nevertheless, FTY720 induces cardiotoxicity at the anti-neoplastic dose. Here, we have developed a series of non-phosphorylable FTY720 analogues as a new therapeutic strategy for AML. Our results show that the lead compound CM-1231 re-activates PP2A by targeting SET-PP2A interaction, inhibiting cell proliferation and promoting apoptosis in AML cell lines and primary patient samples. Notably, CM-1231 did not induce cardiac toxicity, unlike FTY720, in zebrafish models, and reduced the invasion and aggressiveness of AML cells more than FTY720 in zebrafish xenograft models. In conclusion, CM-1231 is safer and more effective than FTY720; therefore, this compound could represent a novel and promising approach for treating AML patients with SET overexpression.


Asunto(s)
Cardiotoxicidad/prevención & control , Proteínas de Unión al ADN/metabolismo , Clorhidrato de Fingolimod/administración & dosificación , Chaperonas de Histonas/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteína Fosfatasa 2/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis/efectos de los fármacos , Cardiotoxicidad/etiología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Clorhidrato de Fingolimod/análogos & derivados , Clorhidrato de Fingolimod/toxicidad , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Unión Proteica/efectos de los fármacos , Pruebas de Toxicidad Aguda , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
8.
Neuropharmacology ; 158: 107701, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31291595

RESUMEN

Multiple system atrophy (MSA) is a fatal demyelinating disorder lacking any disease-modifying therapies. MSA pathology stems from aggregated α-synuclein (aSyn) accumulation in glial cytosolic inclusions of oligodendroglial cell (OLGs), the myelinating cells of brain. In MSA brains and in MSA animal models with aSyn accumulation in OLGs, aberrant expression of brain-derived neurotrophic factor (BDNF) and glial-cell-line-derived neurotrophic factor (GDNF) occur. Nerve growth factor (NGF) expression can also be altered in neurodegenerative diseases. It is unclear if oxidative stress impacts the viability of aSyn-accumulating OLG cells. Here, we show that OLN-93 cells stably expressing human wild type aSyn or the MSA-associated-aSyn-mutants G51D or A53E, are more vulnerable to oxidative stress. In dose response studies we found that OLN-93 cells treated 48 h with 160 nM FTY720 or our new non-immunosuppressive FTY720-C2 or FTY720-Mitoxy derivatives sustained normal viability. Also, FTY720, FTY720-C2, and FTY720-Mitoxy all stimulated NGF expression at 24 h. However only FTY720-Mitoxy also increased BDNF and GDNF mRNA at 24 h, an effect paralleled by increases in histone 3 acetylation and ERK1/2 phosphorylation. Myelin associated glycoprotein (MAG) levels were also increased in OLN-93 cells after 48 h treatment with FTY720-Mitoxy. FTY720, FTY720-C2, and FTY720-Mitoxy all prevented oxidative-stress-associated-cell-death of OLN-93 cells that lack any aSyn expression. However, only FTY720-Mitoxy protected MSA-like aSyn-expressing-OLN-93-cells against oxidative-cell-death. These data identify potent protective effects for FTY720-Mitoxy with regard to trophic factors as well as MAG expression by OLG cells. Testing of FTY720-Mitoxy in mice is thus a judicious next step for neuropharmacological preclinical development.


Asunto(s)
Ceramidas/farmacología , Clorhidrato de Fingolimod/análogos & derivados , Atrofia de Múltiples Sistemas/metabolismo , Oligodendroglía/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología , alfa-Sinucleína/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular , Clorhidrato de Fingolimod/farmacología , Factor Neurotrófico Derivado de la Línea Celular Glial/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Glicoproteína Asociada a Mielina/efectos de los fármacos , Glicoproteína Asociada a Mielina/metabolismo , Factor de Crecimiento Nervioso/efectos de los fármacos , Factor de Crecimiento Nervioso/metabolismo , Oligodendroglía/metabolismo , Ratas , alfa-Sinucleína/metabolismo
9.
J Neurol Sci ; 399: 6-14, 2019 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-30738334

RESUMEN

Subarachnoid hemorrhage (SAH) results in neurological damage, acute cardiac damage and has a high mortality rate. Immunoresponse in the acute phase after SAH plays a key role in mediating vasospasm, edema, inflammation and neuronal damage. The S1P/S1PR pathway impacts multiple cellular functions, exerts anti-inflammatory and anti-apoptotic effects, promotes remyelination, and improves outcome in several central nervous system (CNS) diseases. RP001 hydrochloride is a novel S1PR agonist, which sequesters lymphocytes within their secondary tissues and prevents infiltration of immune cells into the CNS thereby reducing immune response. In this study, we investigated whether RP001 attenuates neuronal injury after SAH by reducing inflammation. S1PRs, specifically S1PR1, 3 not only exerts anti-inflammatory effects, but also decreases heart rate and induces atrioventricular conduction abnormalities. Therefore, we also tested whether RP001 treatment of SAH regulates cardiac functional outcome. Male adult C57BL/6 mice were subjected to SAH, and neurological function tests, echocardiography, and immunohistochemical analysis were performed. SAH induces neurological deficits and acute cardiac dysfunction compared to sham control mice. Treatment of SAH with a low-dose of RP001 induces better neurological outcome and cardiac function compared to a high-dose of RP001. Low-dose-RP001 treatment significantly decreases apoptosis, white matter damage, blood brain barrier permeability, microglial/astrocyte activation, macrophage chemokine protein-1, matrix metalloproteinase-9 and NADPH oxidase-2 expression in the brain compared to SAH control mice. Our findings indicate that low-dose of RP001 alleviates neurological damage after SAH, in part by decreasing neuroinflammation.


Asunto(s)
Encéfalo/efectos de los fármacos , Neuronas/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos , Moduladores de los Receptores de fosfatos y esfingosina 1/uso terapéutico , Receptores de Esfingosina-1-Fosfato/agonistas , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod/análogos & derivados , Frecuencia Cardíaca/efectos de los fármacos , Inflamación/tratamiento farmacológico , Masculino , Ratones , Neuronas/metabolismo , Transducción de Señal/efectos de los fármacos , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología , Hemorragia Subaracnoidea/metabolismo
10.
Pharmazie ; 74(2): 107-110, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30782260

RESUMEN

SYL-927 is a selective sphingosine-1-phosphate receptor 1 (S1P1) agonist for autoimmune diseases. It undergoes phosphorylation to the active SYL-927-P in vivo, which activates S1P1 on lymphocytes, causing lymphopenia by retention of lymphocytes in the lymph nodes. The aim of this study was to identify the involvement of blood cells in the phosphorylation of SYL-927. In addition, pharmacokinetics of SYL-927 and SYL-927-P in blood and plasma were compared in rats. The results demonstrated that SYL-927 can be converted to SYL-927-P in rat blood, but not in rat plasma. However, both rat blood and plasma are capable of dephosphorylating SYL-927-P to SYL-927. SYL-927-P generation and release were observed after incubating SYL-927 with rat and human erythrocytes and platelets. The addition of sphingosine kinases (SPHKs) inhibitors N,N-dimethylsphingosine (DMS) and FTY720 significantly inhibited SYL-927-P generation, indicating the involvement of SPHKs. In addition, SYL-927 and SYL-927-P levels in blood were significantly higher than those in plasma after oral administration of SYL-927 in rats, suggesting the blood cells for the production of SYL-927-P. In summary, the blood cells such as erythrocytes and platelets contribute to the generation and release of SYL-927-P, which is important for maintaining plasma active phosphate levels for prolonged effects.


Asunto(s)
Plaquetas/metabolismo , Eritrocitos/metabolismo , Clorhidrato de Fingolimod/análogos & derivados , Inmunosupresores/sangre , Receptores de Lisoesfingolípidos/agonistas , Administración Oral , Animales , Clorhidrato de Fingolimod/administración & dosificación , Clorhidrato de Fingolimod/sangre , Humanos , Inmunosupresores/administración & dosificación , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/sangre , Ratas , Esfingosina/administración & dosificación , Esfingosina/análogos & derivados , Esfingosina/sangre
11.
Neurosci Lett ; 690: 178-180, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30359694

RESUMEN

In searching for Parkinson's disease (PD) pharmacotherapies we began studying FTY720, a food and drug administration (FDA) approved drug. We also created derivatives, FTY720-C2 and FTY720-Mitoxy, and began assessing them. Here we treated dopaminergic MN9D cells with FTY720s then measured microRNA (miRNA) levels by PCR arrays. We discovered that all three FTY720s increased miR376b-3p, while FTY720-C2 also increased miR-128-3p, miR-146b-5p, miR-7a-5p, and miR-9-5p, and FTY720-Mitoxy also increased miR-30d-5p. Investigations revealed that some miRNAs downregulate alpha-synuclein, while others reduce apoptosis, suggesting that FTY720s may act to reduce synucleinopathy and dopaminergic neuron loss in PD and related disorders.


Asunto(s)
Ceramidas/farmacología , Neuronas Dopaminérgicas/metabolismo , Clorhidrato de Fingolimod/análogos & derivados , Clorhidrato de Fingolimod/farmacología , MicroARNs/metabolismo , Fármacos Neuroprotectores/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Células Cultivadas , Ratones
12.
Eur J Med Chem ; 159: 217-242, 2018 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-30292898

RESUMEN

A series of compounds containing pyrrolidine and pyrrolizidine cores with appended hydrophobic substituents were prepared as constrained analogs of FTY720 and phytosphingosine. The effect of these compounds on the viability of cancer cells, on downregulation of the nutrient transport systems, and on their ability to cause vacuolation was studied. An attempt to inhibit HDACs with some phosphate esters of our analogs was thwarted by our failure to reproduce the reported inhibitory action of FTY720-phosphate.


Asunto(s)
Antineoplásicos/farmacología , Epigénesis Genética/efectos de los fármacos , Clorhidrato de Fingolimod/farmacología , Esfingosina/análogos & derivados , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Clorhidrato de Fingolimod/análogos & derivados , Clorhidrato de Fingolimod/química , Ratones , Estructura Molecular , Esfingosina/síntesis química , Esfingosina/química , Esfingosina/farmacología , Relación Estructura-Actividad
13.
Molecules ; 23(11)2018 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-30355990

RESUMEN

FTY720 inhibits various cancers through PP2A activation. The structure of FTY720 is also used as a basic structure for the design of sphingosine kinase (SK) inhibitors. We have synthesized derivatives using an amide chain in FTY720 with a phenyl backbone, and then compounds were screened by an MTT cell viability assay. The PP2A activity of compound 7 was examined. The phosphorylation levels of AKT and ERK, downstream targets of PP2A, in the presence of compound 7, were determined. Compound 7 may exhibit anticancer effects through PP2A activation rather than the mechanism by inhibition of SK1 in cancer cells. In the docking study of compound 7 and PP2A, the amide chain of compound 7 showed an interaction with Asn61 that was different from FTY720, which is expected to affect the activity of the compound.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Clorhidrato de Fingolimod/síntesis química , Clorhidrato de Fingolimod/farmacología , Proteína Fosfatasa 2/antagonistas & inhibidores , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Clorhidrato de Fingolimod/análogos & derivados , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Relación Estructura-Actividad
14.
Biopharm Drug Dispos ; 39(9): 431-436, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30362120

RESUMEN

SYL-927, a novel and selective S1P1 agonist, is transferred to its active phosphate for the regulation of lymphocyte recirculation. This in vitro metabolism study is to elucidate the P450-mediated oxidation pathway of SYL-927 in human liver microsomes (HLMs). The results demonstrated that the ω-1 hydroxylated metabolite SYL-927-M was formed after incubation of SYL-927 with HLMs. Recombinant human CYP1A1 and CYP2J2 can efficiently catalyse SYL-927-M formation, followed by markedly less substrate conversion with CYP1A2, CYP2C19 and CYP2D6. Inhibition studies with chemical inhibitors and antibodies suggested that arachidonic acid, the substrate of CYP2J2, and CYP2J2-specific antibody effectively inhibited the formation of SYL-927-M in HLMs whereas no significant inhibition was observed with the inhibitors for CYP1A1, CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1 and CYP3A4, demonstrating that CYP2J2 was primarily responsible for the formation of SYL-927-M.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Clorhidrato de Fingolimod/análogos & derivados , Microsomas Hepáticos/enzimología , Oxazoles/metabolismo , Receptores de Esfingosina-1-Fosfato/agonistas , Citocromo P-450 CYP2J2 , Humanos , Hidroxilación , Oxazoles/farmacología
15.
Chem Commun (Camb) ; 54(85): 12002-12005, 2018 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-30221278

RESUMEN

The direct, catalytic vicinal difluorination of terminal alkenes via an I(i)/I(iii) manifold was exploited to install a chiral, hybrid bioisostere of the CF3 and Et groups (BITE) in Gilenya®; the first orally available drug for the clinical management of Multiple Sclerosis (MS). This subtle fluorination pattern allows lipophilicity (log D) to be tempered compared to the corresponding CF3 and Et derivatives (CH2CH3 > CH2CF3 > CHFCH2F).


Asunto(s)
Clorhidrato de Fingolimod/análogos & derivados , Clorhidrato de Fingolimod/química , Inmunosupresores/química , Animales , Estabilidad de Medicamentos , Clorhidrato de Fingolimod/síntesis química , Halogenación , Hepatocitos/efectos de los fármacos , Humanos , Inmunosupresores/síntesis química , Microsomas Hepáticos/metabolismo , Esclerosis Múltiple/tratamiento farmacológico , Ratas
16.
Bioorg Med Chem ; 24(10): 2273-9, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27068143

RESUMEN

SYL927 and SYL930 are selective S1P1 agonists under preclinical development. However, during their pharmacokinetic studies we detected two metabolites in rat blood that were tentatively identified as monohydroxylated metabolites of SYL927 and SYL930 based on LC-MS/MS data. In this study, we designed and synthesized possible monohydroxylated products 6a-e and used them as references to confirm the structures of the two metabolites detected by LC-MS/MS. We also evaluated the in vitro and in vivo biological activities of these two metabolites.


Asunto(s)
Clorhidrato de Fingolimod/análogos & derivados , Clorhidrato de Fingolimod/farmacología , Inmunosupresores/química , Inmunosupresores/farmacología , Receptores de Lisoesfingolípidos/agonistas , Animales , Cromatografía Liquida , Clorhidrato de Fingolimod/administración & dosificación , Hidroxilación , Inmunosupresores/administración & dosificación , Recuento de Linfocitos , Linfocitos/efectos de los fármacos , Ratas Sprague-Dawley , Receptores de Lisoesfingolípidos/inmunología , Espectrometría de Masas en Tándem
17.
Chem Phys Lipids ; 194: 85-93, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26496151

RESUMEN

Effective therapeutic agents are lacking for the prevention and reversal of vascular leak, a frequent pathophysiologic result of inflammatory processes such as acute respiratory distress syndrome (ARDS) and sepsis. We previously demonstrated the potent barrier-enhancing effects of related compounds sphingosine 1-phosphate (S1P), the pharmaceutical agent FTY720, and its analog (S)-FTY720 phosphonate (Tys) in models of inflammatory lung injury. In this study, we characterize additional novel FTY720 analogs for their potential to reduce vascular leak as well as utilize them as tools to better understand the mechanisms by which this class of agents modulates permeability. Transendothelial resistance (TER) and labeled dextran studies demonstrate that (R)-methoxy-FTY720 ((R)-OMe-FTY), (R)/(S)-fluoro-FTY720 (FTY-F), and ß-glucuronide-FTY720 (FTY-G) compounds display in vitro barrier-enhancing properties comparable or superior to FTY720 and S1P. In contrast, the (S)-methoxy-FTY720 ((S)-OMe-FTY) analog disrupts lung endothelial cell (EC) barrier integrity in TER studies in association with actin stress fiber formation and robust intracellular calcium release, but independent of myosin light chain or ERK phosphorylation. Additional mechanistic studies with (R)-OMe-FTY, FTY-F, and FTY-G suggest that lung EC barrier enhancement is mediated through lipid raft signaling, Gi-linked receptor coupling to downstream tyrosine phosphorylation events, and S1PR1-dependent receptor ligation. These results provide important mechanistic insights into modulation of pulmonary vascular barrier function by FTY720-related compounds and highlight common signaling events that may assist the development of novel therapeutic tools in the prevention or reversal of the pulmonary vascular leak that characterizes ARDS.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Clorhidrato de Fingolimod/análogos & derivados , Clorhidrato de Fingolimod/farmacología , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Células Cultivadas , Células Endoteliales/metabolismo , Clorhidrato de Fingolimod/química , Humanos , Permeabilidad/efectos de los fármacos , Arteria Pulmonar/metabolismo , Relación Estructura-Actividad
18.
ACS Chem Biol ; 11(2): 409-14, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26653336

RESUMEN

FTY720 sequesters lymphocytes in secondary lymphoid organs through effects on sphingosine-1-phosphate (S1P) receptors. However, at higher doses than are required for immunosuppression, FTY720 also functions as an anticancer agent in multiple animal models. Our published work indicates that the anticancer effects of FTY720 do not depend on actions at S1P receptors but instead stem from FTY720s ability to restrict access to extracellular nutrients by down-regulating nutrient transporter proteins. This result was significant because S1P receptor activation is responsible for FTY720s dose-limiting toxicity, bradycardia, that prevents its use in cancer patients. Here, we describe diastereomeric and enantiomeric 3- and 4-C-aryl 2-hydroxymethyl pyrrolidines that are more active than the previously known analogues. Of importance is that these compounds fail to activate S1P1 or S1P3 receptors in vivo but retain inhibitory effects on nutrient transporter proteins and anticancer activity in solid tumor xenograft models. Our studies reaffirm that the anticancer activity of FTY720 does not depend upon S1P receptor activation and uphold the promise of using S1P receptor-inactive azacyclic FTY720 analogues in human cancer patients.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Clorhidrato de Fingolimod/análogos & derivados , Clorhidrato de Fingolimod/uso terapéutico , Neoplasias/tratamiento farmacológico , Pirrolidinas/química , Pirrolidinas/uso terapéutico , Animales , Antineoplásicos/farmacología , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Clorhidrato de Fingolimod/farmacología , Humanos , Inmunosupresores/química , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Ratones Desnudos , Neoplasias/metabolismo , Neoplasias/patología , Pirrolidinas/farmacología , Receptores de Lisoesfingolípidos/metabolismo
19.
Chem Phys Lipids ; 191: 16-24, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26272033

RESUMEN

Effective therapeutic agents are lacking for the prevention and reversal of vascular leak, a frequent pathophysiologic result of inflammatory processes such as acute respiratory distress syndrome (ARDS) and sepsis. We previously demonstrated the potent barrier-enhancing effects of related compounds sphingosine 1-phosphate (S1P), the pharmaceutical agent FTY720, and its analog (S)-FTY720 phosphonate (Tys) in models of inflammatory lung injury. In this study, we characterize additional novel FTY720 analogs for their potential to reduce vascular leak as well as utilize them as tools to better understand the mechanisms by which this class of agents modulates permeability. Transendothelial resistance (TER) and labeled dextran studies demonstrate that (R)-methoxy-FTY720 ((R)-OMe-FTY), (R)/(S)-fluoro-FTY720 (FTY-F), and ß-glucuronide-FTY720 (FTY-G) compounds display in vitro barrier-enhancing properties comparable or superior to FTY720 and S1P. In contrast, the (S)-methoxy-FTY720 ((S)-OMe-FTY) analog disrupts lung endothelial cell (EC) barrier integrity in TER studies in association with actin stress fiber formation and robust intracellular calcium release, but independent of myosin light chain or ERK phosphorylation. Additional mechanistic studies with (R)-OMe-FTY, FTY-F, and FTY-G suggest that lung EC barrier enhancement is mediated through lipid raft signaling, Gi-linked receptor coupling to downstream tyrosine phosphorylation events, and S1PR1-dependent receptor ligation. These results provide important mechanistic insights into modulation of pulmonary vascular barrier function by FTY720-related compounds and highlight common signaling events that may assist the development of novel therapeutic tools in the prevention or reversal of the pulmonary vascular leak that characterizes ARDS.


Asunto(s)
Clorhidrato de Fingolimod/análogos & derivados , Calcio/metabolismo , Línea Celular , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Clorhidrato de Fingolimod/farmacología , Fluoruros/química , Glucurónidos/química , Humanos , Lisofosfolípidos/metabolismo , Microscopía Fluorescente , Permeabilidad/efectos de los fármacos , Fosforilación , Arteria Pulmonar/citología , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/metabolismo
20.
Microvasc Res ; 99: 102-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25862132

RESUMEN

RATIONALE: Modulation of pulmonary vascular barrier function is an important clinical goal given the devastating effects of vascular leak in acute lung injury (ALI). We previously demonstrated that FTY720 S-phosphonate (Tys), an analog of sphingosine 1-phosphate (S1P) and FTY720, has more potent pulmonary barrier protective effects than these agents in vitro and in mouse models of ALI. Tys preserves expression of the barrier-promoting S1P1 receptor (S1PR1), whereas S1P and FTY720 induce its ubiquitination and degradation. Here we further characterize the novel barrier promoting effects of Tys in cultured human pulmonary endothelial cells (EC). METHODS/RESULTS: In human lung EC, Tys significantly increased peripheral redistribution of adherens junction proteins VE-cadherin and ß-catenin and tight junction protein ZO-1. Inhibition of VE-cadherin with blocking antibody significantly attenuated Tys-induced transendothelial resistance (TER) elevation, while ZO-1 siRNA partially inhibited this elevation. Tys significantly increased focal adhesion formation and phosphorylation of focal adhesion kinase (FAK). Pharmacologic inhibition of FAK significantly attenuated Tys-induced TER elevation. Tys significantly increased phosphorylation and peripheral redistribution of the actin-binding protein, cortactin, while cortactin siRNA partially attenuated Tys-induced TER elevation. Although Tys significantly increased phosphorylation of Akt and GSK3ß, neither PI3 kinase nor GSK3ß inhibition altered Tys-induced TER elevation. Tys significantly increased Rac1 activity, while inhibition of Rac1 activity significantly attenuated Tys-induced VE-cadherin redistribution and TER elevation. CONCLUSION: Junctional complex, focal adhesion rearrangement and Rac1 activation play critical roles in Tys-mediated barrier protection in pulmonary EC. These results provide mechanistic insights into the effects of this potential ALI therapy.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Clorhidrato de Fingolimod/análogos & derivados , Clorhidrato de Fingolimod/uso terapéutico , Pulmón/irrigación sanguínea , Organofosfonatos/uso terapéutico , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/patología , Antígenos CD/metabolismo , Antígenos Nucleares/metabolismo , Cadherinas/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Adhesiones Focales/metabolismo , Humanos , Inmunosupresores/uso terapéutico , Pulmón/efectos de los fármacos , Lisofosfolípidos/química , Microscopía Fluorescente , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/química , Factores de Transcripción/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Proteína de Unión al GTP rac1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA