Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Front Immunol ; 15: 1321309, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38469297

RESUMO

Background: The thymus plays a central role in shaping human immune function. A mechanistic, quantitative description of immune cell dynamics and thymic output under homeostatic conditions and various patho-physiological scenarios are of particular interest in drug development applications, e.g., in the identification of potential therapeutic targets and selection of lead drug candidates against infectious diseases. Methods: We here developed an integrative mathematical model of thymocyte dynamics in human. It incorporates mechanistic features of thymocyte homeostasis as well as spatial constraints of the thymus and considerations of age-dependent involution. All model parameter estimates were obtained based on published physiological data of thymocyte dynamics and thymus properties in mouse and human. We performed model sensitivity analyses to reveal potential therapeutic targets through an identification of processes critically affecting thymic function; we further explored differences in thymic function across healthy subjects, multiple sclerosis patients, and patients on fingolimod treatment. Results: We found thymic function to be most impacted by the egress, proliferation, differentiation and death rates of those thymocytes which are most differentiated. Model predictions also showed that the clinically observed decrease in relapse risk with age, in multiple sclerosis patients who would have discontinued fingolimod therapy, can be explained mechanistically by decreased thymic output with age. Moreover, we quantified the effects of fingolimod treatment duration on thymic output. Conclusions: In summary, the proposed model accurately describes, in mechanistic terms, thymic output as a function of age. It may be further used to perform predictive simulations of clinically relevant scenarios which combine specific patho-physiological conditions and pharmacological interventions of interest.


Assuntos
Esclerose Múltipla , Timócitos , Humanos , Camundongos , Animais , Timócitos/metabolismo , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Cloridrato de Fingolimode/metabolismo , Timo , Diferenciação Celular , Esclerose Múltipla/metabolismo
2.
Cell Rep ; 42(12): 113545, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38064339

RESUMO

Vitamin B12 (B12) deficiency causes neurological manifestations resembling multiple sclerosis (MS); however, a molecular explanation for the similarity is unknown. FTY720 (fingolimod) is a sphingosine 1-phosphate (S1P) receptor modulator and sphingosine analog approved for MS therapy that can functionally antagonize S1P1. Here, we report that FTY720 suppresses neuroinflammation by functionally and physically regulating the B12 pathways. Genetic and pharmacological S1P1 inhibition upregulates a transcobalamin 2 (TCN2)-B12 receptor, CD320, in immediate-early astrocytes (ieAstrocytes; a c-Fos-activated astrocyte subset that tracks with experimental autoimmune encephalomyelitis [EAE] severity). CD320 is also reduced in MS plaques. Deficiency of CD320 or dietary B12 restriction worsens EAE and eliminates FTY720's efficacy while concomitantly downregulating type I interferon signaling. TCN2 functions as a chaperone for FTY720 and sphingosine, whose complex induces astrocytic CD320 internalization, suggesting a delivery mechanism of FTY720/sphingosine via the TCN2-CD320 pathway. Taken together, the B12-TCN2-CD320 pathway is essential for the mechanism of action of FTY720.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Animais , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Cloridrato de Fingolimode/metabolismo , Astrócitos/metabolismo , Esfingosina/metabolismo , Vitamina B 12/farmacologia , Vitamina B 12/uso terapêutico , Vitamina B 12/metabolismo , Transcobalaminas/metabolismo , Transcobalaminas/uso terapêutico , Propilenoglicóis/metabolismo , Propilenoglicóis/farmacologia , Propilenoglicóis/uso terapêutico , Vitaminas , Imunossupressores/farmacologia , Receptores de Lisoesfingolipídeo/metabolismo
3.
Mol Immunol ; 160: 55-66, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37379683

RESUMO

Lupus nephritis (LN) is a common complication of systemic lupus erythematosus (SLE) as well as the leading cause of mortality in patients. Previous studies revealed that S1P level is elevated in plasma samples of SLE patients and murine lupus models. FTY720, targeting S1P receptors, exhibited therapeutic effects in improving the nephritis symptoms of lupus mouse models. However, few studies have discussed the potential relevance of S1P/S1PR to the pathogenesis of LN. Macrophages have been shown to be an important causative agent of renal inflammation, while the pro-inflammatory M1-type promotes kidney injury and inflammation during LN. Importantly, macrophages express various S1P receptors, and how they respond to S1P in the setting of LN remains unclear. Therefore, we examined the level of S1P in the lupus MRL/lpr mice and explored the ensuing interaction of macrophages and S1P. We found that S1P level was elevated in the MRL/lpr mice with a subsequent enhancement of the S1PR1 expression, and blocking S1PR1 by FTY720, the nephritis symptoms of MRL/lpr mice were improved. Mechanistically, we demonstrated that elevated S1P level increase the M1-type macrophage accumulation. And the in-vitro studies proved that S1P/S1PR1 was involved in the promotion of macrophage polarization towards M1 type through activation of NLRP3 inflammasome. These findings confer a novel role to macrophage S1PR1 and provide a new perspective for targeting S1P during LN.


Assuntos
Lúpus Eritematoso Sistêmico , Nefrite Lúpica , Animais , Camundongos , Cloridrato de Fingolimode/metabolismo , Cloridrato de Fingolimode/uso terapêutico , Inflamassomos/metabolismo , Inflamação/patologia , Lúpus Eritematoso Sistêmico/metabolismo , Nefrite Lúpica/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos MRL lpr , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Transdução de Sinais , Receptores de Esfingosina-1-Fosfato/metabolismo
4.
Neurotoxicology ; 96: 197-206, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37160207

RESUMO

Gulf War Illness (GWI) is an unrelenting multi-symptom illness with chronic central nervous system and peripheral pathology affecting veterans from the 1991 Gulf War and for which effective treatment is lacking. An increasing number of studies indicate that persistent neuroinflammation is likely the underlying cause of cognitive and mood dysfunction that affects veterans with GWI. We have previously reported that fingolimod, a drug approved for the treatment of relapsing-remitting multiple sclerosis, decreases neuroinflammation and improves cognition in a mouse model of Alzheimer's disease. In this study, we investigated the effect of fingolimod treatment on cognition and neuroinflammation in a mouse model of GWI. We exposed C57BL/6 J male mice to GWI-related chemicals pyridostigmine bromide, DEET, and permethrin, and to mild restraint stress for 28 days (GWI mice). Control mice were exposed to the chemicals' vehicle only. Starting 3 months post-exposure, half of the GWI mice and control mice were orally treated with fingolimod (1 mg/kg/day) for 1 month, and the other half were left untreated. Decreased memory on the Morris water maze test was detected in GWI mice compared to control mice and was reversed by fingolimod treatment. Immunohistochemical analysis of brain sections with antibodies to Iba1 and GFAP revealed that GWI mice had increased microglia activation in the hippocampal dentate gyrus, but no difference in reactive astrocytes was detected. The increased activation of microglia in GWI mice was decreased to the level in control mice by treatment with fingolimod. No effect of fingolimod treatment on gliosis in control mice was detected. To explore the signaling pathways by which decreased memory and increased neuroinflammation in GWI may be protected by fingolimod, we investigated the involvement of the inflammatory signaling pathways of protein kinase R (PKR) in the cerebral cortex of these mice. We found increased phosphorylation of PKR in the brain of GWI mice compared to controls, as well as increased phosphorylation of its most recognized downstream effectors: the α subunit of eukaryotic initiation factor 2 (eIF2α), IκB kinase (IKK), and the p65 subunit of nuclear factor-κB (NFκB-p65). Furthermore, we found that the increased phosphorylation level of these three proteins were suppressed in GWI mice treated with fingolimod. These results suggest that activation of PKR and NFκB signaling may be important for the regulation of cognition and neuroinflammation in the GWI condition and that fingolimod, a drug already approved for human use, may be a potential candidate for the treatment of GWI.


Assuntos
Cloridrato de Fingolimode , Síndrome do Golfo Pérsico , Animais , Masculino , Camundongos , Amnésia/metabolismo , Modelos Animais de Doenças , Cloridrato de Fingolimode/uso terapêutico , Cloridrato de Fingolimode/metabolismo , Cloridrato de Fingolimode/farmacologia , Guerra do Golfo , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , Camundongos Endogâmicos C57BL , Microglia , Doenças Neuroinflamatórias , NF-kappa B/metabolismo , Síndrome do Golfo Pérsico/induzido quimicamente , Síndrome do Golfo Pérsico/tratamento farmacológico , Síndrome do Golfo Pérsico/metabolismo , Proteínas Quinases/metabolismo , Proteínas Quinases/farmacologia , Proteínas Quinases/uso terapêutico , Brometo de Piridostigmina/uso terapêutico , Brometo de Piridostigmina/farmacologia
5.
Behav Brain Res ; 446: 114415, 2023 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-36997095

RESUMO

Sphingosine-1-phosphate (S1P) is emerging as a crucial sphingolipid modulating neuroinflammation and cognition. S1P levels in the brain have been found to be decreased in cognitive impairment. S1P lyase (S1PL) is the key enzyme in metabolizing S1P and has been implicated in neuroinflammation. This study evaluated the effect of S1PL inhibition on cognition in type 2 diabetic mice. Fingolimod (0.5 mg/kg and 1 mg/kg) rescued cognition in high-fat diet and streptozotocin-induced diabetic mice, as evident in the Y maze and passive avoidance test. We further evaluated the effect of fingolimod on the activation of microglia in the pre-frontal cortex (PFC) and hippocampus of diabetic mice. Our study revealed that fingolimod inhibited S1PL and promoted anti-inflammatory microglia in both PFC and hippocampus of diabetic mice as it increased Ym-1 and arginase-1. The levels of p53 and apoptotic proteins (Bax and caspase-3) were elevated in the PFC and hippocampus of type 2 diabetic mice which fingolimod reversed. The underlying mechanism promoting anti-inflammatory microglial phenotype was also explored in this study. TIGAR, TP53-associated glycolysis and apoptosis regulator, is known to foster anti-inflammatory microglia and was found to be downregulated in the brain of type 2 diabetic mice. S1PL inhibition decreased the levels of p53 and promoted TIGAR, thereby increasing anti-inflammatory microglial phenotype and inhibiting apoptosis in the brain of diabetic mice. Our study reveals that S1PL inhibition could be beneficial in mitigating cognitive deficits in diabetic mice.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Camundongos , Animais , Esfingosina/farmacologia , Esfingosina/metabolismo , Cloridrato de Fingolimode/metabolismo , Cloridrato de Fingolimode/farmacologia , Microglia , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/farmacologia , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Doenças Neuroinflamatórias , Cognição , Diabetes Mellitus Tipo 2/metabolismo , Fosfatos/metabolismo , Fosfatos/farmacologia , Monoéster Fosfórico Hidrolases/metabolismo , Monoéster Fosfórico Hidrolases/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo
6.
J Control Release ; 355: 248-258, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36736432

RESUMO

Glioblastoma (GBM) is the most invasive brain tumor and remains lack of effective treatment. The existence of blood-brain tumor barrier (BBTB) constitutes the greatest barrier to non-invasive delivery of therapeutic agents to tumors in the brain. Here, we propose a novel approach to specifically modulate BBTB and deliver magnetic hyperthermia in a systemic delivery mode for the treatment of GBM. BBTB modulation is achieved by targeted delivering fingolimod to brain tumor region via dual redox responsive PCL-SeSe-PEG (poly (ε-caprolactone)-diselenium-poly (ethylene glycol)) polymeric nanocarrier. As an antagonist of sphingosine 1-phosphate receptor-1 (S1P1), fingolimod potently inhibits the barrier function of BBB by blocking the binding of sphingosine 1-phosphate (S1P) to S1P1 in endothelial cells. We found that the modulated BBTB showed slight expression level of tight junction proteins, allowing efficient accumulation of zinc- and cobalt- doped iron oxide nanoclusters (ZnCoFe NCs) with enhanced magnetothermal conversion efficiency into tumor tissues through the paracellular pathway. As a result, the co-delivery of heat shock protein 70 inhibitor VER-155008 with ZnCoFe NCs could realize synergistic magnetic hyperthermia effects upon exposure to an alternating current magnetic field (ACMF) in both GL261 and U87 brain tumor models. This modulation approach brings new ideas for the treatment of central nervous system diseases that require delivery of therapeutic agents across the blood-brain barrier (BBB).


Assuntos
Neoplasias Encefálicas , Glioblastoma , Hipertermia Induzida , Humanos , Barreira Hematoencefálica/metabolismo , Cloridrato de Fingolimode/metabolismo , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Células Endoteliais/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Fenômenos Magnéticos
7.
Int J Mol Sci ; 23(18)2022 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-36142246

RESUMO

Transdifferentiation of Schwann cells is essential for functional peripheral nerve regeneration after injury. By activating a repair program, Schwann cells promote functional axonal regeneration and remyelination. However, chronic denervation, aging, metabolic diseases, or chronic inflammatory processes reduce the transdifferentiation capacity and thus diminish peripheral nerve repair. It was recently described that the sphingosine-1-phosphate receptor (S1PR) agonist Fingolimod enhances the Schwann cell repair phenotype by activation of dedifferentiation markers and concomitant release of trophic factors resulting in enhanced neurite growth. Since Fingolimod targets four out of five S1PRs (S1P1, S1P3-5) possibly leading to non-specific adverse effects, identification of the main receptor(s) responsible for the observed phenotypic changes is mandatory for future specific treatment approaches. Our experiments revealed that S1P3 dominates and that along with S1P1 acts as the responsible receptor for Schwann cell transdifferentiation as revealed by the combinatory application of specific agonists and antagonists. Targeting both receptors reduced the expression of myelin-associated genes, increased PDGF-BB representing enhanced trophic factor expression likely to result from c-Jun induction. Furthermore, we demonstrated that S1P4 and S1P5 play only a minor role in the adaptation of the repair phenotype. In conclusion, modulation of S1P1 and S1P3 could be effective to enhance the Schwann cell repair phenotype and thus stimulate proper nerve repair.


Assuntos
Cloridrato de Fingolimode , Células de Schwann , Becaplermina/metabolismo , Cloridrato de Fingolimode/metabolismo , Cloridrato de Fingolimode/farmacologia , Regeneração Nervosa/fisiologia , Fenótipo , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Células de Schwann/metabolismo , Receptores de Esfingosina-1-Fosfato
8.
Proteomics ; 22(19-20): e2100247, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35866514

RESUMO

Fingolimod (FTY720) is an oral drug approved by the Food and Drug Administration (FDA) for management of multiple sclerosis (MS) symptoms, which has also shown beneficial effects against Alzheimer's (AD) and Parkinson's (PD) diseases pathologies. Although an extensive effort has been made to identify mechanisms underpinning its therapeutic effects, much remains unknown. Here, we investigated Fingolimod induced proteome changes in the cerebellum (CB) and frontal cortex (FC) regions of the brain which are known to be severely affected in MS, using a tandem mass tag (TMT) isobaric labeling-based quantitative mass-spectrometric approach to investigate the mechanism of action of Fingolimod. This study identified 6749 and 6319 proteins in CB and FC, respectively, and returned 2609 and 3086 differentially expressed proteins in mouse CB and FC, respectively, between Fingolimod treated and control groups. Subsequent bioinformatics analyses indicated a metabolic reprogramming in both brain regions of the Fingolimod treated group, where oxidative phosphorylation was upregulated while glycolysis and pentose phosphate pathway were downregulated. In addition, modulation of neuroinflammation in the Fingolimod treated group was indicated by upregulation of retrograde endocannabinoid signaling and autophagy pathways, and downregulation of neuroinflammation related pathways including neutrophil degranulation and the IL-12 mediated signaling pathway. Our findings suggest that Fingolimod may exert its protective effects on the brain by inducing metabolic reprogramming and neuroinflammation pathway modulation.


Assuntos
Cloridrato de Fingolimode , Esclerose Múltipla , Animais , Camundongos , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/metabolismo , Cloridrato de Fingolimode/uso terapêutico , Proteoma/metabolismo , Endocanabinoides/metabolismo , Encéfalo/metabolismo , Esclerose Múltipla/metabolismo , Metabolismo Energético , Autofagia , Interleucina-12/metabolismo
9.
Biomed Pharmacother ; 152: 113240, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35689862

RESUMO

The protection mediated by the bioactive sphingolipid sphingosine-1-phosphate (S1P) declines during Alzheimer's disease (AD) progression, especially in patients carrying the apolipoprotein E ε4 (APOE4) isoform. The drug FTY720 mimics S1P bioactivity, but its efficacy in treating AD is unclear. Two doses of FTY720 (0.1 mg / kg and 0.5 mg / kg daily) were given by oral gavage for 15 weeks to transgenic mouse models of familial AD carrying human apolipoprotein E (APOE) APOE3 (E3FAD) or APOE4 (E4FAD). After 12 weeks of treatment, animals were subjected to behavioral tests for memory, locomotion, and anxiety. Blood was withdrawn at different time points and brains were collected for sphingolipids analysis by mass spectrometry, gene expression by RT-PCR and Aß quantification by ELISA. We discovered that low levels of S1P in the plasma is associated with a higher probability of failing the memory test and that FTY720 prevents memory impairments in E4FAD. The beneficial effect of FTY720 was induced by a shift of the sphingolipid metabolism in the brain towards a lower production of toxic metabolites, like ceramide d18:1/16:0 and d18:1/22:0, and reduction of amyloid-ß burden and inflammation. In conclusion, we provide further evidence of the druggability of the sphingolipid system in AD.


Assuntos
Doença de Alzheimer , Apolipoproteína E4 , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/prevenção & controle , Animais , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteína E4/uso terapêutico , Encéfalo/metabolismo , Ceramidas/metabolismo , Modelos Animais de Doenças , Cloridrato de Fingolimode/metabolismo , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Humanos , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , Transtornos da Memória/prevenção & controle , Camundongos , Esfingolipídeos/metabolismo
10.
J Mol Cell Cardiol ; 164: 110-125, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34774547

RESUMO

Effects of hypertrophic challenge on small-conductance, Ca2+-activated K+(SK2) channel expression were explored in intact murine hearts, isolated ventricular myocytes and neonatal rat cardiomyocytes (NRCMs). An established experimental platform applied angiotensin II (Ang II) challenge in the presence and absence of reduced p21-activated kinase (PAK1) (PAK1cko vs. PAK1f/f, or shRNA-PAK1 interference) expression. SK2 current contributions were detected through their sensitivity to apamin block. Ang II treatment increased such SK2 contributions to optically mapped action potential durations (APD80) and their heterogeneity, and to patch-clamp currents. Such changes were accentuated in PAK1cko compared to PAK1f/f, intact hearts and isolated cardiomyocytes. They paralleled increased histological and echocardiographic hypertrophic indices, reduced cardiac contractility, and increased SK2 protein expression, changes similarly greater with PAK1cko than PAK1f/f. In NRCMs, Ang II challenge replicated such increases in apamin-sensitive SK patch clamp currents as well as in real-time PCR and western blot measures of SK2 mRNA and protein expression and cell hypertrophy. Furthermore, the latter were enhanced by shRNA-PAK1 interference and mitigated by the PAK1 agonist FTY720. Increased CaMKII and CREB phosphorylation accompanied these effects. These were rescued by both FTY720 as well as the CaMKII inhibitor KN93, but not its inactive analogue KN92. Such CREB then specifically bound to the KCNN2 promoter sequence in luciferase assays. These findings associate Ang II induced hypertrophy with increased SK2 expression brought about by a CaMKII/CREB signaling convergent with the PAK1 pathway thence upregulating the KCNN2 promoter activity. SK2 may then influence cardiac electrophysiology under conditions of cardiac hypertrophy and failure.


Assuntos
Angiotensina II , Quinases Ativadas por p21 , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Animais , Apamina/metabolismo , Apamina/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cardiomegalia/metabolismo , Cloridrato de Fingolimode/metabolismo , Cloridrato de Fingolimode/farmacologia , Camundongos , Miócitos Cardíacos/metabolismo , Proteínas Serina-Treonina Quinases , RNA Interferente Pequeno/metabolismo , Ratos , Regulação para Cima , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo , Quinases Ativadas por p21/farmacologia
11.
Cell Transplant ; 30: 9636897211052975, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34662222

RESUMO

Autologous fat grafting (AFG) is widely regarded as an important method for breast reconstruction after mastectomy among breast cancer (BC) patients. FTY720 has been proved to affect macrophage polarization and improve the sensitivity of postoperative BC treatment. This study aimed to explore FTY720 function and underlying mechanism in fat transplantation. The C57BL/6 J mice that received AFG were randomly divided into two groups treated with saline and FTY720, respectively. The fat graft samples were obtained at week 1, 2, 4, and 12 post-transplantation. Graft volumes, graft structures, M2 macrophages, and STAT3 protein expression were estimated by histological examination, immunofluorescence, flow cytometry, and western blot, respectively. In vitro, mouse preadipocytes were stimulated with FTY720 treated-M2 macrophages conditioned medium (FTY720-M2-CM) to evaluate the adipogenesis effect. The level of adipogenic mRNA expression in preadipocytes was detected by RT-PCR. The in vivo results showed that FTY720 treatment significantly enhanced the fat graft retention, structure integrity, and neovascularization, indicating the potential of FTY720 in improving graft survival. The histology results showed more polarized M2 macrophage presented in the FTY720 group. In the in vitro assay, after FTY720-M2-CM treatment, the 3T3-L1 preadipocytes showed the increased triglyceride content and adipogenic mRNA expression, including FABP4, C/EBP-α, Adipoq, and PPARγ. Furthermore, FTY720 treatment up-regulated the expression level of M2 biomarker CD206, Arg-1, Fizz-1, which could be weakened by the STAT3 inhibitor. Together, this study confirmed the potential efficacy of FTY720 in improving graft survival in the AFG model, possibly mediated by polarizing macrophages to M2 type through activating the STAT3 pathway.


Assuntos
Tecido Adiposo/metabolismo , Cloridrato de Fingolimode/metabolismo , Macrófagos/metabolismo , Fator de Transcrição STAT3/metabolismo , Transplante Autólogo/métodos , Transplantes/transplante , Animais , Feminino , Humanos , Camundongos , Transdução de Sinais
12.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34502385

RESUMO

Erythropoietin (Epo) is the critical hormone for erythropoiesis. In adults, Epo is mainly produced by a subset of interstitial fibroblasts in the kidney, with minor amounts being produced in the liver and the brain. In this study, we used the immortalized renal interstitial fibroblast cell line FAIK F3-5 to investigate the ability of the bioactive sphingolipid sphingosine 1-phosphate (S1P) to stimulate Epo production and to reveal the mechanism involved. Stimulation of cells with exogenous S1P under normoxic conditions (21% O2) led to a dose-dependent increase in Epo mRNA and protein levels and subsequent release of Epo into the medium. S1P also enhanced the stabilization of HIF-2α, a key transcription factor for Epo expression. S1P-stimulated Epo mRNA and protein expression was abolished by HIF-2α mRNA knockdown or by the HIF-2 inhibitor compound 2. Furthermore, the approved S1P receptor modulator FTY720, and its active form FTY720-phosphate, both exerted a similar effect on Epo expression as S1P. The effect of S1P on Epo was antagonized by the selective S1P1 and S1P3 antagonists NIBR-0213 and TY-52156, but not by the S1P2 antagonist JTE-013. Moreover, inhibitors of the classical MAPK/ERK, the p38-MAPK, and inhibitors of protein kinase (PK) C and D all blocked the effect of S1P on Epo expression. Finally, the S1P and FTY720 effects were recapitulated in the Epo-producing human neuroblastoma cell line Kelly, suggesting that S1P receptor-dependent Epo synthesis is of general relevance and not species-specific. In summary, these data suggest that, in renal interstitial fibroblasts, which are the primary source of plasma Epo, S1P1 and 3 receptor activation upregulates Epo under normoxic conditions. This may have a therapeutic impact on disease situations such as chronic kidney disease, where Epo production is impaired, causing anemia, but it may also have therapeutic value as Epo can mediate additional tissue-protective effects in various organs.


Assuntos
Eritropoetina/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Linhagem Celular , Células Cultivadas , Eritropoese , Eritropoetina/fisiologia , Fibroblastos/metabolismo , Cloridrato de Fingolimode/metabolismo , Humanos , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Rim/metabolismo , Lisofosfolipídeos/metabolismo , Camundongos , Ligação Proteica , Receptores de Lisoesfingolipídeo/metabolismo , Insuficiência Renal Crônica/metabolismo , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/fisiologia
13.
Cell Signal ; 76: 109806, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33035646

RESUMO

The sphingosine kinases, SK1 and SK2, catalyse the formation of the bioactive signalling lipid, sphingosine 1-phosphate (S1P), from sphingosine. SK1 and SK2 differ in their subcellular localisation, trafficking and regulation, but the isoforms are also distinct in their selectivity toward naturally occurring and synthetic ligands as substrates and inhibitors. To date, only the structure of SK1 has been determined, and a structural basis for selectivity differences in substrate handling by SK2 has yet to be established. Here we present a structural rationale, based on homology modelling and ligand docking, to account for the capacity of SK2, but not SK1, to efficiently process the pharmacologically active substances, fingolimod (FTY720) and safingol, as substrates. We propose that two key residue differences in hSK2 (Ser305/Thr584 in place of Ala175/Ala339 in hSK1) facilitate conformational switching in the lipid head group anchor residue, Asp308 (corresponding to Asp178 in hSK1), to accommodate substrate diversity for SK2. Our analysis accounts for the contrasting behaviour of fingolimod and safingol as non-turnover inhibitors of SK1, but substrates for SK2, and the observed stereoselectivity for phosphorylation of the pro-S hydroxymethyl group of fingolimod to generate (S)-FTY720-P in vivo. We also rationalise why methylation of the pro-R hydroxymethyl of FTY720 switches the behaviour of the resulting compound, (R)-FTY720 methyl ether (ROMe), to SK2-selective inhibition. Whilst the pharmacological significance of (S)-FTY720-P is firmly established, as the active principle of fingolimod in treating relapsing-remitting multiple sclerosis, the potential importance of SK-mediated phosphorylation of other substrates, such as safingol and non-canonical naturally occuring substrates such as (4E,nZ)-sphingadienes, is less widely appreciated. Thus, the contribution of SK2-derived safingol 1-phosphate to the anti-cancer activity of safingol should be considered. Similarly, the biological role of sphingadiene 1-phosphates derived from plant-based dietary sphingadienes, which we also show here are substrates for both SK1 and SK2, merits investigation.


Assuntos
Inibidores Enzimáticos , Cloridrato de Fingolimode/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Moduladores do Receptor de Esfingosina 1 Fosfato/metabolismo , Esfingosina/análogos & derivados , Animais , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Ligantes , Esfingosina/metabolismo
14.
Int J Mol Sci ; 21(18)2020 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-32911595

RESUMO

During heart transplantation, donor heart leads to reduced oxygen supply resulting in low level of high energy phosphate (HEP) reserves in cardiomyocyte. Lower HEP is one of the underlying reasons of cell death due to ischemia. In this study we investigated the role of Fingolimod (FTY720) in heart transplantation ischemia. Eight groups of Sprague-Dawley rats (n = 5 for each subgroup) were made, A1 and C1 were given FTY720 1 mg/kg while B1 and D1 were given normal saline. The hearts were implanted into another set of similar rats after preservation period of 1 h at 4-8 °C. Significantly higher Left ventricular systolic pressure (LVSP), dP/dT maximum (p < 0.05), dP/dT minimum (p < 0.05) were recorded in the FTY720 treated group after 24 h of reperfusion while after 1 h of reperfusion, there were no significant differences in LVSP, maximum and negative dP/dT, and Left ventricular end diastolic pressure (LVEDP) between the control and the FTY720-treated transplant groups. Coronary blood flow (CBF) was enhanced (p < 0.05) in the FTY720 treated group after 1 and 24 h. ATP p < 0.001, p < 0.05 at 1 and 24 h, ADP p < 0.001, p > 0.05 at 1 and 24 h, and phosphocreatine p < 0.05, p > 0.05 at 1 and 24 h were better preserved by FTY720 treatment as compared to control group. The study concluded that pretreatment of grafted hearts with FTY720 improved hemodynamics, CBF, high energy phosphate reserves, reduces the peroxynitrite level and poly (ADP ribose) polymerase (PARP) inhibition that prevents ischemia-reperfusion injury.


Assuntos
Circulação Coronária/efeitos dos fármacos , Cloridrato de Fingolimode/farmacologia , Coração/fisiopatologia , Animais , Doença da Artéria Coronariana/tratamento farmacológico , Doença da Artéria Coronariana/fisiopatologia , Cloridrato de Fingolimode/metabolismo , Coração/efeitos dos fármacos , Transplante de Coração/métodos , Isquemia Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Fosfatos , Poli(ADP-Ribose) Polimerases/metabolismo , Ratos , Ratos Sprague-Dawley
15.
Clin Pharmacol Drug Dev ; 9(5): 610-620, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32468719

RESUMO

Fingolimod is indicated for the treatment of patients with the relapsing-remitting form of multiple sclerosis. The primary study objective was to evaluate the bioequivalence of a test formulation, 0.5 mg fingolimod HCl capsule (Lebrina, Asofarma Sociedad Anónima Industrial y Comercial, Argentina) relative to a reference formulation, 0.5 mg fingolimod capsule (Gilenya, Novartis Pharmaceutical, Australia). In a single-center, randomized, single-dose, single-blinded, 2-way crossover study, 33 New Zealand healthy subjects of both sexes were enrolled to receive a 0.5-mg dose of 3 capsules of each fingolimod formulation under fasting conditions, with a 42-day washout period between administrations. Additional pharmacokinetic information regarding its main active metabolite, fingolimod phosphate, was also provided. The point estimate and 90% confidence intervals of the ratios of maximum concentration and area under the plasma concentration-time curve from time 0 to 72 hours were 99.07 (95.83-102.41) and 97.64 (95.33-100.00) for fingolimod, and 95.60 (90.95-100.49) and 98.54 (96.19-100.96), for fingolimod phosphate. Primary parameters, maximum concentration and area under the plasma concentration-time curve from time 0 to 72 hours for fingolimod and fingolimod phosphate were found to have no significant difference when test and reference formulations were compared. Fingolimod and fingolimod phosphate of both formulations were within the accepted 90% confidence interval limits of 80.00% and 125.00%. No significant differences between the test and reference drug products were detected in any of the pharmacokinetic parameters estimated. Notwithstanding the primary conclusion of bioequivalence is focused on the measurement of the parent compound, compliance with the same criteria by the active metabolite reinforces the comparability between the pharmacokinetic profiles of both formulations (ClinicalTrials.gov Identifier: NCT03757338).


Assuntos
Família 4 do Citocromo P450/metabolismo , Composição de Medicamentos/métodos , Cloridrato de Fingolimode/farmacocinética , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Moduladores do Receptor de Esfingosina 1 Fosfato/farmacocinética , Administração Oral , Adulto , Área Sob a Curva , Índice de Massa Corporal , Estudos Cross-Over , Composição de Medicamentos/estatística & dados numéricos , Jejum/metabolismo , Feminino , Cloridrato de Fingolimode/administração & dosagem , Cloridrato de Fingolimode/sangue , Cloridrato de Fingolimode/metabolismo , Voluntários Saudáveis/estatística & dados numéricos , Humanos , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla Recidivante-Remitente/sangue , Nova Zelândia/epidemiologia , Moduladores do Receptor de Esfingosina 1 Fosfato/administração & dosagem , Moduladores do Receptor de Esfingosina 1 Fosfato/sangue , Moduladores do Receptor de Esfingosina 1 Fosfato/metabolismo , Equivalência Terapêutica
16.
PLoS One ; 15(4): e0230846, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32240213

RESUMO

PURPOSE: The objective of this study was to characterize the demographic and clinical profile of RRMS patients receiving fingolimod in Spain, and to evaluate drug effectiveness and safety in clinical practice. METHODS: This observational, retrospective, multicentre, nationwide study was performed at 56 Spanish hospitals and involved 804 RRMS patients who received oral fingolimod (0.5 mg) since November 2011, with a minimum follow-up of 12 months. RESULTS: The mean annualized relapse rate (ARR) in the year before fingolimod was 1.08 and the median EDSS was 3; patients were exposed to fingolimod for 2.2 years as average; regarding magnetic resonance imaging (MRI) activity, more than half of the patients had >20 lesions at baseline. Patients were previously treated with first-line injectable DMTs (60.3%), or natalizumab (31.3%), and 8.3% were naïve patients. Overall, the ARR significantly decreased to 0.28, 0.22 and 0.17 (74.1%, 79.7% and 83.5% of relative reduction, respectively) after 12, 24 and 36 months of treatment, P<0.001. The ARR of patients who switched from natalizumab to fingolimod was stable over the study. Most of the patients (88.7%) were free from confirmed disability and MRI activity (67.3%) after 24 months. The persistence after 12 months on fingolimod was 93.9%. CONCLUSIONS: The subgroups of patients analysed showed differential baseline demographic and clinical characteristics. The analysis of patients who received fingolimod in routine clinical practice confirmed adequate efficacy and safety, even for long-term treatment. The present data also confirmed the positive benefit/risk balance with fingolimod in real-world clinical practice setting.


Assuntos
Cloridrato de Fingolimode/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Adulto , Idoso , Pessoas com Deficiência , Feminino , Cloridrato de Fingolimode/metabolismo , Humanos , Imunossupressores/uso terapêutico , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Natalizumab/uso terapêutico , Recidiva , Estudos Retrospectivos , Espanha
17.
Transl Stroke Res ; 11(5): 1103-1116, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32103462

RESUMO

Injury due to brain ischemia followed by reperfusion (I/R) may be an important therapeutic target in the era of thrombectomy. FTY720, a widely known sphingosine-1-phosphate receptor agonist, exerts various neuroprotective effects. The aim of this study was to examine the protective effect of FTY720 with respect to I/R injury, especially focusing on blood-brain barrier (BBB) protection and anti-inflammatory effects. Male rats were subjected to transient ischemia and administered vehicle or 0.5 or 1.5 mg/kg of FTY720 immediately before reperfusion. Positron emission tomography (PET) with [18F]DPA-714 was performed 2 and 9 days after the insult to serially monitor neuroinflammation. Bovine and rat brain microvascular endothelial cells (MVECs) were also subjected to oxygen-glucose deprivation (OGD) and reperfusion, and administered FTY720, phosphorylated-FTY720 (FTY720-P), or their inhibitor. FTY720 dose-dependently reduced cell death, the infarct size, cell death including apoptosis, and inflammation. It also ameliorated BBB disruption and neurological deficits compared to in the vehicle group. PET indicated that FTY720 significantly inhibited the worsening of inflammation in later stages. FTY720-P significantly prevented the intracellular redistribution of tight junction proteins but did not increase their mRNA expression. These results suggest that FTY720 can ameliorate I/R injury by protecting the BBB and regulating neuroinflammation.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Cloridrato de Fingolimode/farmacologia , Inflamação/tratamento farmacológico , Organofosfatos/farmacologia , Esfingosina/análogos & derivados , Animais , Apoptose/efeitos dos fármacos , Células Endoteliais/metabolismo , Cloridrato de Fingolimode/metabolismo , Ratos Sprague-Dawley , Esfingosina/farmacologia , Proteínas de Junções Íntimas/metabolismo
19.
Brain Res Bull ; 153: 109-121, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31442591

RESUMO

It has been shown that fingolimod (FTY720) possesses beneficial effects on remyelination in the central nervous system (CNS). In this study, the effects of FTY720 and sodium valproate (VPA) as histone deacetylase inhibitor (HDAC) on the conductivity of visual signals, extent of demyelination area, glial activation, and expression levels of HDAC1and S1PR1 have been evaluated in the optic chiasm of lysolecithin (LPC)-induced demyelination model. In order to produce this demyelination model, LPC (1%, 2 µL) was injected into the rat optic chiasm. Latency of visual waves was measured by visual evoked potential (VEP) recording. The extent of demyelination area and level of glial activation were assessed using immunostaining. Gene expression analysis was performed to evaluate the expression levels of HDAC1, S1PR1, Olig2, and MBP in the optic chiasm. Analysis of electrophysiological data showed that LPC administration increased the latency of visual signals. FTY720 improved the functional recovery of the visual pathway and reduced the level of glial activation in the optic chiasm. FTY720 enhanced myelin repair and up-regulated the expression levels of Olig2 and MBP. Additionally, the expression levels of HDAC1 and S1PR1 were significantly reduced in animals treated with FTY720. In contrast to FTY720 treated animals, administration of VPA could not significantly improve the functional recovery of optic pathway following LPC injection. Cumulatively, the results of the present study demonstrate that FTY720 application improves the functional recovery of the optic pathway by reducing demyelination levels, amelioration of glial activation, and down-regulating of S1PR1 and HDAC1.


Assuntos
Cloridrato de Fingolimode/farmacologia , Quiasma Óptico/efeitos dos fármacos , Remielinização/efeitos dos fármacos , Animais , Doenças Desmielinizantes/induzido quimicamente , Modelos Animais de Doenças , Cloridrato de Fingolimode/metabolismo , Histona Desacetilase 1/metabolismo , Lisofosfatidilcolinas , Masculino , Esclerose Múltipla , Bainha de Mielina/fisiologia , Oligodendroglia/efeitos dos fármacos , Quiasma Óptico/metabolismo , Ratos , Ratos Wistar , Recuperação de Função Fisiológica , Receptores de Esfingosina-1-Fosfato/metabolismo , Ácido Valproico/farmacologia
20.
Brain Res ; 1720: 146294, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31201815

RESUMO

Oligodendrocytes (OLGs) support neuronal system and have crucial roles for brain homeostasis. As the renewal and regeneration of OLGs derived from oligodendrocyte precursor cells (OPCs) are inhibited by various pathological conditions, the restoration of impaired oligodendrogenesis is a therapeutic strategy for OLG-related diseases such as subcortical ischemic vascular dementia (SIVD). Fingolimod (FTY720), a drug for multiple sclerosis, is reported to elicit a cytoprotective effect on OPCs in vitro. However, the effects of fingolimod against ischemia-induced suppression of OPC differentiation remain unknown. Hence, the purpose of this study was to investigate the effectiveness of fingolimod against ischemia-induced suppression of oligodendrogenesis. For the in vitro experiments, primary rat cultured OPCs were incubated with a non-lethal concentration of CoCl2 to induce chemical hypoxic conditions and were treated with or without fingolimod-phosphate. We found that low concentration fingolimod-phosphate directly rescued ischemia-induced suppression of OPC differentiation via the phosphoinositide 3-kinase-Akt pathway. For the in vivo experiments, we used a mouse model of SIVD generated by bilateral common carotid artery stenosis. On day 28 after surgery, fingolimod ameliorated ischemia-induced demyelination and promoted oligodendrogenesis under prolonged cerebral hypoperfusion. The present study demonstrates that fingolimod can promote oligodendrogenesis under ischemic conditions and may be a therapeutic candidate for SIVD.


Assuntos
Cloridrato de Fingolimode/farmacologia , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Oligodendroglia/metabolismo , Animais , Isquemia Encefálica/patologia , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Feminino , Cloridrato de Fingolimode/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/fisiologia , Células Precursoras de Oligodendrócitos/metabolismo , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Substância Branca/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...