Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Mol Pharm ; 21(7): 3310-3320, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38856116

RESUMEN

Siponimod is a promising agent for the inhibition of ocular neovascularization in diabetic retinopathy and age-related macular degeneration. Siponimod's development for ophthalmological application is hindered by the limited information available on the drug's solubility, stability, ocular pharmacokinetics (PK), and toxicity in vivo. In this study, we investigated the aqueous stability of siponimod under stress conditions (up to 60 °C) and its degradation behavior in solution. Additionally, siponimod's ocular PK and toxicity were investigated using intravitreal injection of two different doses (either 1300 or 6500 ng) in an albino rabbit model. Siponimod concentration was quantified in the extracted vitreous, and the PK parameters were calculated. The drug half-life after administration of the low and high doses was 2.8 and 3.9 h, respectively. The data obtained in vivo was used to test the ability of published in silico models to predict siponimod's PK accurately. Two models that correlated siponimod's molecular descriptors with its elimination from the vitreous closely predicted the half-life. Furthermore, 24 h and 7 days after intravitreal injections, the retinas showed no signs of toxicity. This study provides important information necessary for the formulation and development of siponimod for ophthalmologic applications. The short half-life of siponimod necessitates the development of a sustained drug delivery system to maintain therapeutic concentrations over an extended period, while the lack of short-term ocular toxicity observed in the retinas of siponimod-treated rabbits supports possible clinical use.


Asunto(s)
Azetidinas , Inyecciones Intravítreas , Animales , Conejos , Azetidinas/farmacocinética , Azetidinas/administración & dosificación , Semivida , Cuerpo Vítreo/efectos de los fármacos , Cuerpo Vítreo/metabolismo , Masculino , Retina/efectos de los fármacos , Retina/metabolismo , Ojo/efectos de los fármacos , Ojo/metabolismo , Retinopatía Diabética/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacocinética , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/toxicidad , Solubilidad , Degeneración Macular/tratamiento farmacológico , Compuestos de Bencilo
2.
Nat Rev Neurol ; 19(8): 489-505, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37438431

RESUMEN

Migraine is a leading cause of disability in more than one billion people worldwide, yet it remains universally underappreciated, even by individuals with the condition. Among other shortcomings, current treatments (often repurposed agents) have limited efficacy and potential adverse effects, leading to low treatment adherence. After the introduction of agents that target the calcitonin gene-related peptide pathway, another new drug class, the ditans - a group of selective serotonin 5-HT1F receptor agonists - has just reached the international market. Here, we review preclinical studies from the late 1990s and more recent clinical research that contributed to the development of the ditans and led to their approval for acute migraine treatment by the US Food and Drug Administration and the European Medicines Agency.


Asunto(s)
Trastornos Migrañosos , Receptores de Serotonina , Humanos , Receptores de Serotonina/uso terapéutico , Trastornos Migrañosos/tratamiento farmacológico , Trastornos Migrañosos/inducido químicamente , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina/farmacología , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina/uso terapéutico , Péptido Relacionado con Gen de Calcitonina , Receptor de Serotonina 5-HT1F
3.
Eur J Immunol ; 53(9): e2350370, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37366289

RESUMEN

Fingolimod has generally shown neuroprotective effects in stroke models. Here, we tested the hypothesis that fingolimod modulates T-cell cytokine production towards a regulatory phenotype. Second, we investigated how fingolimod altered the Treg suppressive function and the sensitivity of effector T cells to regulation. Mice that had underwent the permanent electrocoagulation of the left middle cerebral artery received saline or fingolimod (0.5 mg/kg) daily for 10-days post-ischaemia. Fingolimod improved neurobehavioural recovery compared to saline control and increased Treg frequency in the periphery and brain. Tregs from fingolimod-treated animals had a higher expression of CCR8. Fingolimod increased the frequencies of CD4+ IL-10+ , CD4+ IFN-γ+ and CD4+ IL-10+ IFN-γ+ cells in spleen and blood, and CD4+ IL-17+ cells in the spleen, with only minor effects on CD8+ T-cell cytokine production. Treg from post-ischaemic mice had reduced suppressive function compared to Treg from non-ischaemic mice. Fingolimod treatment rescued this function against saline-treated but not fingolimod-treated CD4+ effector T cells. In conclusion, fingolimod seems to improve the suppressive function of Treg post-stroke while also increasing the resistance of CD4+ effector cells to this suppression. Fingolimod's capacity to increase both effector and regulatory functions may explain the lack of consistent improvement in functional recovery in experimental brain ischaemia.


Asunto(s)
Isquemia Encefálica , Clorhidrato de Fingolimod , Ratones , Animales , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Linfocitos T Reguladores/metabolismo , Interleucina-10/metabolismo , Expresión Génica , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo
4.
J Headache Pain ; 24(1): 62, 2023 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-37237336

RESUMEN

BACKGROUND AND AIMS: Cortical spreading depression (CSD), a transient neuronal and glial depolarization that propagates slowly across the cerebral cortex, is the putative electrophysiological event underlying migraine aura and a headache trigger. Migraine is three times more prevalent in women than men, linked to circulating female hormones. High estrogen levels or estrogen withdrawal may be a migraine trigger for many women. We, therefore, aimed to examine whether sex, gonadectomy, and female hormone supplementation and withdrawal affect the susceptibility to CSD. METHODS: To determine CSD susceptibility, we recorded the frequency of CSDs triggered during 2-h topical KCl application in intact or gonadectomized female and male rats, without or with estradiol or progesterone supplementation via daily intraperitoneal injections. Estrogen or progesterone treatment followed by withdrawal was studied in a separate cohort. To take the first step towards identifying potential mechanisms, we studied glutamate and GABAA receptor binding using autoradiography. RESULTS: The CSD frequency in intact female rats was higher than intact male and ovariectomized rats. We did not detect a change in CSD frequency during different stages of the estrous cycle in intact females. Daily estrogen injections for three weeks did not change CSD frequency. However, one-week estrogen withdrawal after two weeks of treatment significantly increased CSD frequency compared with the vehicle group in gonadectomized females. The same protocol of estrogen treatment and withdrawal was ineffective in gonadectomized males. In contrast to estrogen, daily progesterone injections for three weeks elevated CSD susceptibility, and one-week withdrawal after two weeks of treatment partially normalized this effect. Autoradiography did not reveal significant changes in glutamate or GABAA receptor binding density after estrogen treatment and withdrawal. CONCLUSIONS: These data suggest that females are more susceptible to CSD, and sexual dimorphism is abrogated by gonadectomy. Moreover, estrogen withdrawal after prolonged daily treatment enhances CSD susceptibility. These findings may have implications for estrogen-withdrawal migraine, although the latter tends to be without aura.


Asunto(s)
Depresión de Propagación Cortical , Trastornos Migrañosos , Ratas , Femenino , Masculino , Animales , Depresión de Propagación Cortical/fisiología , Progesterona/farmacología , Receptores de GABA-A , Estrógenos/farmacología , Glutamatos/farmacología
5.
J Pharmacol Exp Ther ; 386(2): 224-241, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37188532

RESUMEN

Sphingosine-1-phosphate (S1P) receptors control endothelial cell proliferation, migration, and survival. Evidence of the ability of S1P receptor modulators to influence multiple endothelial cell functions suggests their potential use for antiangiogenic effect. The main purpose of our study was to investigate the potential of siponimod for the inhibition of ocular angiogenesis in vitro and in vivo. We investigated the effects of siponimod on the metabolic activity (thiazolyl blue tetrazolium bromide assay), cell toxicity (lactate dehydrogenase release), basal proliferation and growth factor-induced proliferation (bromodeoxyuridine assay), and migration (transwell migration assay) of human umbilical vein endothelial cells (HUVEC) and retinal microvascular endothelial cells (HRMEC). The effects of siponimod on HRMEC monolayer integrity, barrier function under basal conditions, and tumor necrosis factor alpha (TNF-α)-induced disruption were assessed using the transendothelial electrical resistance and fluorescein isothiocyanate-dextran permeability assays. Siponimod's effect on TNF-α-induced distribution of barrier proteins in HRMEC was investigated using immunofluorescence. Finally, the effect of siponimod on ocular neovascularization in vivo was assessed using suture-induced corneal neovascularization in albino rabbits. Our results show that siponimod did not affect endothelial cell proliferation or metabolic activity but significantly inhibited endothelial cell migration, increased HRMEC barrier integrity, and reduced TNF-α-induced barrier disruption. Siponimod also protected against TNF-α-induced disruption of claudin-5, zonula occludens-1, and vascular endothelial-cadherin in HRMEC. These actions are mainly mediated by sphingosine-1-phosphate receptor 1 modulation. Finally, siponimod prevented the progression of suture-induced corneal neovascularization in albino rabbits. In conclusion, the effects of siponimod on various processes known to be involved in angiogenesis support its therapeutic potential in disorders associated with ocular neovascularization. SIGNIFICANCE STATEMENT: Siponimod is an extensively characterized sphingosine-1-phosphate receptor modulator already approved for the treatment of multiple sclerosis. It inhibited retinal endothelial cell migration, potentiated endothelial barrier function, protected against tumor necrosis factor alpha-induced barrier disruption, and also inhibited suture-induced corneal neovascularization in rabbits. These results support its use for a novel therapeutic indication in the management of ocular neovascular diseases.


Asunto(s)
Neovascularización de la Córnea , Factor de Necrosis Tumoral alfa , Animales , Humanos , Conejos , Retina , Neovascularización Patológica , Células Endoteliales de la Vena Umbilical Humana , Células Cultivadas
6.
Stroke ; 53(12): 3741-3750, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36252110

RESUMEN

BACKGROUND: It has been reported that the S1P (sphingosine 1-phosphate) receptor modulator fingolimod reduces infarction in rodent models of stroke. Recent studies have suggested that circadian rhythms affect stroke and neuroprotection. Therefore, this study revisited the use of fingolimod in mouse focal cerebral ischemia to test the hypothesis that efficacy might depend on whether experiments were performed during the inactive sleep or active wake phases of the circadian cycle. METHODS: Two different stroke models were implemented in male C57Bl/6 mice-transient middle cerebral artery occlusion and permanent distal middle cerebral artery occlusion. Occlusion occurred either during inactive or active circadian phases. Mice were treated with 1 mg/kg fingolimod at 30- or 60-minute postocclusion and 1 day later for permanent and transient middle cerebral artery occlusion, respectively. Infarct volume, brain swelling, hemorrhagic transformation, and behavioral outcome were assessed at 2 or 3 days poststroke. Three independent experiments were performed in 2 different laboratories. RESULTS: Fingolimod decreased peripheral lymphocyte number in naive mice, as expected. However, it did not significantly affect infarct volume, brain swelling, hemorrhagic transformation, or behavioral outcome at 2 or 3 days after transient or permanent focal cerebral ischemia during inactive or active circadian phases of stroke onset. CONCLUSIONS: Outcomes were not improved by fingolimod in either transient or permanent focal cerebral ischemia during both active and inactive circadian phases. These negative findings suggest that further testing of fingolimod in clinical trials may not be warranted unless translational studies can identify factors associated with fingolimod's efficacy or lack thereof.


Asunto(s)
Edema Encefálico , Isquemia Encefálica , Accidente Cerebrovascular , Animales , Ratones , Masculino , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Edema Encefálico/tratamiento farmacológico , Esfingosina , Accidente Cerebrovascular/tratamiento farmacológico , Ratones Endogámicos C57BL , Hemorragia/tratamiento farmacológico , Modelos Animales de Enfermedad
7.
Brain Behav Immun Health ; 25: 100497, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36120102

RESUMEN

Background: The well-characterised role of the immune system in acute ischaemic stroke has prompted the search for immunomodulatory therapies. Pregnancy-specific glycoproteins (PSGs) are a group of proteins synthesised by placental trophoblasts which show immunomodulatory properties. The aim of this study was to determine whether a proposed PSG1-based therapeutic enhanced recovery in a mouse model of brain ischaemia and to explore possible immunomodulatory effects. Methods: Mice underwent permanent electrocoagulation of the left middle cerebral artery (pMCAO). They received saline (n = 20) or recombinant pregnancy-specific glycoprotein-1-alpha "fused" to the Fc domain of IgG1 (rPSG1-Fc) (100 µg) (n = 22) at 1 h post-ischaemia. At 3 and 5 days post-ischaemia, neurobehavioural recovery was assessed by the grid-walking test. At 5 days post-ischaemia, lesion size was determined by NeuN staining. Peripheral T cell populations were quantified via flow cytometry. Immunohistochemistry was used to quantify ICAM-1 expression and FoxP3+ cell infiltration in the ischaemic brain. Immunofluorescence was employed to determine microglial activation status via Iba-1 staining.Results: rPSG1-Fc significantly enhanced performance in the grid-walking test at 3 and 5 days post-ischaemia. No effect on infarct size was observed. A significant increase in circulating CD4+ FoxP3+ cells and brain-infiltrating FoxP3+ cells was noted in rPSG1-Fc-treated mice. Among CD4+ cells, rPSG1-Fc enhanced the expression of IL-10 in spleen, blood, draining lymph nodes, and non-draining lymph nodes, while downregulating IFN-γ and IL-17 in spleen and blood. A similar cytokine expression pattern was observed in CD8+ cells. rPSG1-Fc reduced activated microglia in the infarct core. Conclusion: The administration of rPSG1-Fc improved functional recovery in post-ischaemic mice without impacting infarct size. Improved outcome was associated with a modulation of the cytokine-secreting phenotype of CD4+ and CD8+ T cells towards a more regulatory phenotype, as well as reduced activation of microglia. This establishes proof-of-concept of rPSG1-Fc as a potential stroke immunotherapy.

8.
Front Pharmacol ; 13: 920449, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35910379

RESUMEN

Preclinical data indicate that fingolimod improves outcome post-ischaemia. This study used a rigorous study design in normal male C57BL/6JOlaHsd mice and in mice with common stroke comorbidities to further evaluate the translational potential of fingolimod. Stroke was induced via middle cerebral artery electrocoagulation in 8-9-week old mice (young mice), 18 month old mice (aged mice), and in high-fat diet-fed 22-week old ApoE-/- mice (hyperlipidaemic mice). Recovery was evaluated using motor behavioural tests 3 and 7 days after stroke. Tissue damage was evaluated at 7 days. A lower dose of fingolimod, 0.5 mg/kg, but not 1 mg/kg, increased lesion size but decreased ipsilateral brain atrophy in younger mice, without an effect on behavioural outcomes. Fingolimod-treated aged mice showed a significant improvement over saline-treated mice in the foot fault test at 7 days. Fingolimod-treated hyperlipidaemic mice showed a decreased infarct size but no difference in behavioural performance. Increasing fingolimod treatment time to 10 days showed no benefit in young mice. Pooled data showed that fingolimod improved performance in the foot fault test. Flow cytometry studies showed that fingolimod had marked effects on T cell frequencies in various tissues. The results show that the effects of fingolimod in stroke are less robust than the existing literature might indicate and may depend on the inflammatory status of the animals.

9.
Adv Drug Deliv Rev ; 187: 114342, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35569559

RESUMEN

There is an increasing momentum in research and pharmaceutical industry communities to design sustained, non-invasive delivery systems to treat chronic neovascular ocular diseases that affect the posterior segment of the eye including age-related macular degeneration and diabetic retinopathy. Current treatments include VEGF blockers, which have revolutionized the standard of care for patients, but their maximum therapeutic benefit is hampered by the need for recurrent and invasive administration procedures. Currently approved delivery systems intended to address these limitations exploit polymer technology to regulate drug release in a sustained manner. Here, we critically review sustained drug delivery approaches for the treatment of chronic neovascular diseases affecting the ocular posterior segment, with a special emphasis on novel and polymeric technologies spanning the spectrum of preclinical and clinical investigation, and those approved for treatment. The mechanism by which each formulation imparts sustained release, the impact of formulation characteristics on release and foreign body reaction, and special considerations related to the translation of these systems are discussed.


Asunto(s)
Retinopatía Diabética , Degeneración Macular , Retinopatía Diabética/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Ojo , Humanos , Degeneración Macular/tratamiento farmacológico , Polímeros/uso terapéutico
10.
Br J Ophthalmol ; 106(9): 1187-1195, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-33962970

RESUMEN

Neovascular ocular diseases (such as age-related macular degeneration, diabetic retinopathy and retinal vein occlusion) are characterised by common pathological processes that contribute to disease progression. These include angiogenesis, oedema, inflammation, cell death and fibrosis. Currently available therapies target the effects of vascular endothelial growth factor (VEGF), the main mediator of pathological angiogenesis. Unfortunately, VEGF blockers are expensive biological therapeutics that necessitate frequent intravitreal administration and are associated with multiple adverse effects. Thus, alternative treatment options associated with fewer side effects are required for disease management. This review introduces sphingosine 1-phosphate (S1P) as a potential pharmacological target for the treatment of neovascular ocular pathologies. S1P is a sphingolipid mediator that controls cellular growth, differentiation, survival and death. S1P actions are mediated by five G protein-coupled receptors (S1P1-5 receptors) which are abundantly expressed in all retinal and subretinal structures. The action of S1P on S1P1 receptors can reduce angiogenesis, increase endothelium integrity, reduce photoreceptor apoptosis and protect the retina against neurodegeneration. Conversely, S1P2 receptor signalling can increase neovascularisation, disrupt endothelial junctions, stimulate VEGF release, and induce retinal cell apoptosis and degeneration of neural retina. The aim of this review is to thoroughly discuss the role of S1P and its different receptor subtypes in angiogenesis, inflammation, apoptosis and fibrosis in order to determine which of these S1P-mediated processes may be targeted therapeutically.


Asunto(s)
Retinopatía Diabética , Factor A de Crecimiento Endotelial Vascular , Fibrosis , Humanos , Inflamación , Lisofosfolípidos , Neovascularización Patológica , Esfingosina/análogos & derivados , Factores de Crecimiento Endotelial Vascular
11.
Data Brief ; 36: 107146, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34095389

RESUMEN

In this work, the sphingosine-1-phosphate receptor modulator fingolimod was assessed as a preclinical candidate for the treatment of acute ischaemic stroke according to the Stroke Therapy Academic Industry Roundtable (STAIR) preclinical recommendations. Young (15-17 weeks), aged (72-73 weeks), and ApoE-/- mice (20-21 weeks) fed a high fat diet (all C57BL/6 mice) underwent permanent electrocoagulation of the left middle cerebral artery. Mice received either saline or fingolimod (0.5 mg/kg or 1 mg/kg) at 2-, 24-, and 48-hours post-ischaemia via intraperitoneal (i.p.) injection. Another cohort of young mice (8-9 and 17-19 weeks) received short-term (5 days) or long-term (10 days) fingolimod (0.5 mg/kg) treatment in a treatment duration study. For young, aged, and ApoE-/- mice, motor behavioural tests (cylinder and grid-walking) were performed at days 0, 3, and 7 post-ischaemia to evaluate neurobehavioural recovery. In the treatment duration study, the grid-walking test was performed at days 0, 2, 5 and 10 post-ischaemia. Brain tissue sections were stained with haematoxylin and eosin (H&E), and NeuN to quantify tissue damage. Flow cytometry was used to quantify T cell populations in blood, spleen, and lymph nodes. The data presented in this article improves our understanding of the potential neuroprotective and immunomodulatory effects of fingolimod in a mouse model of brain ischaemia. Such data may be significant in the design of future preclinical and clinical stroke studies for fingolimod.

12.
J Neuroinflammation ; 18(1): 37, 2021 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-33516262

RESUMEN

BACKGROUND: The role of the immune system in stroke is well-recognised. Fingolimod, an immunomodulatory agent licensed for the management of relapsing-remitting multiple sclerosis, has been shown to provide benefit in rodent models of stroke. Its mechanism of action, however, remains unclear. We hypothesised fingolimod increases the number and/or function of regulatory T cells (Treg), a lymphocyte population which promotes stroke recovery. The primary aim of this study was to rigorously investigate the effect of fingolimod on Tregs in a mouse model of brain ischaemia. The effect of fingolimod in mice with common stroke-related comorbidities (ageing and hypercholesteremia) was also investigated. METHODS: Young (15-17 weeks), aged C57BL/6 mice (72-73 weeks), and ApoE-/- mice fed a high-fat diet (20-21 weeks) underwent permanent electrocoagulation of the left middle cerebral artery. Mice received either saline or fingolimod (0.5 mg/kg or 1 mg/kg) at 2, 24, and 48 h post-ischaemia via intraperitoneal injection. Another cohort of young mice (8-9, 17-19 weeks) received short-term (5 days) or long-term (10 days) fingolimod (0.5 mg/kg) treatment. Flow cytometry was used to quantify Tregs in blood, spleen, and lymph nodes. Immunohistochemistry was used to quantify FoxP3+ cell infiltration into the ischaemic brain. RESULTS: Fingolimod significantly increased the frequency of Tregs within the CD4+ T cell population in blood and spleen post-ischaemia in all three mouse cohorts compared to untreated ischemic mice. The highest splenic Treg frequency in fingolimod-treated mice was observed in ApoE-/- mice (9.32 ± 1.73% vs. 7.8 ± 3.01% in young, 6.09 ± 1.64% in aged mice). The highest circulating Treg frequency was also noted in ApoE-/- mice (8.39 ± 3.26% vs. 5.43 ± 2.74% in young, 4.56 ± 1.60% in aged mice). Fingolimod significantly increased the number of FoxP3+ cells in the infarct core of all mice. The most pronounced effects were seen when mice were treated for 10 days post-ischaemia. CONCLUSIONS: Fingolimod increases Treg frequency in spleen and blood post-ischaemia and enhances the number of FoxP3+ cells in the ischaemic brain. The effect of fingolimod on this regulatory cell population may underlie its neuroprotective activity and could be exploited as part of future stroke therapy.


Asunto(s)
Envejecimiento/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod/farmacología , Inmunosupresores/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Envejecimiento/inmunología , Animales , Isquemia Encefálica/inmunología , Dieta Alta en Grasa/efectos adversos , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Inmunosupresores/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/inmunología
13.
Int J Pharm ; 590: 119956, 2020 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-33035608

RESUMEN

Sphingosine 1-phosphate (S1P) receptor modulators can influence bone regeneration owing to their positive impact on osteoblast differentiation and neovascularisation. While previous studies have utilised non-specific S1P and fingolimod, this study aims to design and characterise a controlled release vehicle to deliver the specific S1P1 & 5 receptor modulator siponimod and test its effectiveness in rat critical cranial defects. Electrospun scaffolds of poly lactide-co-glycolide (PLGA) were loaded with siponimod at drug:polymer mass ratios of 0.5:100 to 2:100. Where indicated, collagen was co-spun at a collagen:polymer mass ratio of 2:100. Thereafter, scaffolds underwent in vitro physicochemical characterisation and in vivo assessment using a rat cranial defect model. Drug-loaded scaffolds showed controlled release of siponimod, -cytocompatibility with endothelial and osteoblast cells in vitro, and furthermore, showed that released siponimod stimulated osteoblast differentiation and endothelial cell migration. The in vivo cranial defect repair study showed regeneration was occurring in the defect, although there was no significant difference in the extent of mineralisation between scaffold experimental groups. To our knowledge, this is the first study investigating siponimod in bone regeneration. In vitro studies confirm a positive impact on key cells involved in bone regeneration, however, the scaffolds did not result in significant repair of critical cranial defects.


Asunto(s)
Polímeros , Andamios del Tejido , Animales , Azetidinas , Compuestos de Bencilo , Regeneración Ósea , Ratas
14.
Neuropharmacology ; 181: 108326, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-32966847

RESUMEN

We have previously shown that sphingosine kinase 2 (SPK2) interacts with Bcl-2 via its BH3 domain, activating autophagy by inducing the dissociation of Beclin-1/Bcl-2 complexes, and that a TAT-SPK2 peptide containing the BH3 domain of SPK2 protects neurons against ischemic injury. The goals of the present study were to establish the functional significance of these findings, by testing whether TAT-SPK2 was effective in a mouse model of ischemic stroke, and to explore potential underlying mechanisms. Mice were administered with TAT-SPK2 by intraperitoneal injection before or after transient middle cerebral artery occlusion (tMCAO). Infarct volume, neurological deficit and brain water content were assessed 24 h after reperfusion. Mitophagy inhibitor Mdivi-1 and BNIP3 siRNAs were used to examine the involvement of BNIP3-dependent mitophagy in the neuroprotection of TAT-SPK2. Mitophagy was quantified by immunoblotting, immunofluorescence and electron microscopy. The interaction between TAT-SPK2 and Bcl-2, Bcl-2 and BNIP3 was detected by co-immunoprecipitation. In the tMCAO model, pre-treatment with TAT-SPK2 significantly reduced infarct volume, improved neurological function and decreased brain edema. Neuroprotection by TAT-SPK2 was still seen when the peptide was administered 3 h after reperfusion. TAT-SPK2 also significantly improved functional recovery and reduced long-term brain atrophy of the ischemic hemisphere 30 days after administration. Our studies further showed that TAT-SPK2 directly binds to Bcl-2 and disrupts Bcl-2/Beclin-1 or Bcl-2/BNIP3 complexes to induce mitophagy. These results suggest that TAT-SPK2 protects neurons against ischemia reperfusion injury by activating BNIP3-mediated mitophagy. Agents exploiting this molecular mechanism are potential candidates for the treatment of ischemic stroke.


Asunto(s)
Productos del Gen tat/farmacología , Proteínas de la Membrana/agonistas , Proteínas Mitocondriales/agonistas , Mitofagia/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Péptidos/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/farmacología , Daño por Reperfusión/prevención & control , Animales , Autofagia , Beclina-1 , Edema Encefálico/prevención & control , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos ICR , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , ARN Interferente Pequeño/farmacología
15.
Eur J Pharmacol ; 882: 173262, 2020 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-32534075

RESUMEN

The repair of critical bone defects remains a significant therapeutic challenge. While the implantation of drug-eluting scaffolds is an option, a drug with the optimal pharmacological properties has not yet been identified. Agents acting at sphingosine 1-phosphate (S1P) receptors have been considered, but those investigated so far do not discriminate between the five known S1P receptors. This work was undertaken to investigate the potential of the specific S1P1/5 modulator siponimod as a bone regenerative agent, by testing in vitro its effect on cell types critical to the bone regeneration process. hFOB osteoblasts and HUVEC endothelial cells were treated with siponimod and other S1P receptor modulators and investigated for changes in intracellular cyclic AMP content, viability, proliferation, differentiation, attachment and cellular motility. Siponimod showed no effect on the viability and proliferation of osteoblasts and endothelial cells, but increased osteoblast differentiation (as shown by increased alkaline phosphatase activity). Furthermore, siponimod significantly increased endothelial cell motility in scratch and transwell migration assays. These effects on osteoblast differentiation and endothelial cell migration suggest that siponimod may be a potential agent for the stimulation of localised differentiation of osteoblasts in critical bone defects.


Asunto(s)
Azetidinas/farmacología , Compuestos de Bencilo/farmacología , Regeneración Ósea/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología , Fenómenos Fisiológicos Celulares/efectos de los fármacos , Células Cultivadas , AMP Cíclico/metabolismo , Clorhidrato de Fingolimod/farmacología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Osteoblastos/fisiología
16.
Front Pharmacol ; 11: 613103, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33488389

RESUMEN

Intracerebral haemorrhage (ICH) has no specific treatment, but accounts for up to 15% of all strokes and has the highest mortality. Fingolimod (FTY720) is an immunomodulator approved for the management of multiple sclerosis, with abundant evidence of efficacy in experimental ischemic stroke, and more limited evidence in experimental ICH. The goal of this study was to confirm the efficacy of fingolimod in experimental ICH using rigorous and statistically well-powered studies. ICH was induced in C57BL/6JOlaHsd male and female mice by intrastriatal bacterial collagenase injection. Fingolimod (0.5 mg/kg) or saline was administered intraperitoneally after 0.5, 24 and 72 h, in a randomized and blinded manner. Functional improvement with cylinder, wire hanging, and foot fault tests was evaluated one and two weeks later. Lesion volume and hemispheric atrophy were quantified at the 14-day endpoint. There was a higher mortality in saline-treated females compared to fingolimod-treated females and saline-treated males. There was no treatment- or gender-related difference in the behavioural tests. Histological outcome measures did not differ between any of the groups. These results, contrasting with those of previous studies of fingolimod in experimental ICH, emphasize the importance of rigorous testing of this agent in models more representative of the clinical situation.

17.
Front Pharmacol ; 10: 630, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31281252

RESUMEN

The role of immunity in all stages of stroke is increasingly being recognized, from the pathogenesis of risk factors to tissue repair, leading to the investigation of a range of immunomodulatory therapies. In the acute phase of stroke, proposed therapies include drugs targeting pro-inflammatory cytokines, matrix metalloproteinases, and leukocyte infiltration, with a key objective to reduce initial brain cell toxicity. Systemically, the early stages of stroke are also characterized by stroke-induced immunosuppression, where downregulation of host defences predisposes patients to infection. Therefore, strategies to modulate innate immunity post-stroke have garnered greater attention. A complementary objective is to reduce longer-term sequelae by focusing on adaptive immunity. Following stroke onset, the integrity of the blood-brain barrier is compromised, exposing central nervous system (CNS) antigens to systemic adaptive immune recognition, potentially inducing autoimmunity. Some pre-clinical efforts have been made to tolerize the immune system to CNS antigens pre-stroke. Separately, immune cell populations that exhibit a regulatory phenotype (T- and B- regulatory cells) have been shown to ameliorate post-stroke inflammation and contribute to tissue repair. Cell-based therapies, established in oncology and transplantation, could become a strategy to treat the acute and chronic stages of stroke. Furthermore, a role for the gut microbiota in ischaemic injury has received attention. Finally, the immune system may play a role in remote ischaemic preconditioning-mediated neuroprotection against stroke. The development of stroke therapies involving organs distant to the infarct site, therefore, should not be overlooked. This review will discuss the immune mechanisms of various therapeutic strategies, surveying published data and discussing more theoretical mechanisms of action that have yet to be exploited.

18.
Immunol Cell Biol ; 97(1): 5-16, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30022515

RESUMEN

Stroke is a major cause of morbidity and mortality worldwide. Despite the intensive search for new therapies, hundreds of agents targeting various pathophysiological mechanisms have failed clinical trials, and the thrombolytic agent tissue plasminogen activator is currently the only FDA-approved medication for the treatment of acute ischemic stroke. The immune system is involved in all stages of stroke, from the pathogenesis of risk factors to neurotoxicity, to tissue remodeling and repair. There is a bidirectional interaction between the brain and the immune system, with stroke-induced immunosuppression and subsequent infection a principal source of patient mortality. Newer work also points to a role for the gut microbiota in the immune response to stroke, while clinical sequelae such as dementia might now also be explained in immune terms. However, the exact roles of innate and adaptive components have not been fully elucidated, with studies reporting both detrimental and beneficial functions. Time is a key determinant in defining whether immunity and inflammation are neuroprotective or neurotoxic. The local inflammatory milieu also has a clear influence on many proposed treatments. This review examines the individual components of the immune response to stroke, highlighting the most promising future stroke immunotherapies.


Asunto(s)
Inmunoterapia , Accidente Cerebrovascular/inmunología , Inmunidad Adaptativa , Microbioma Gastrointestinal/inmunología , Humanos , Inmunidad Innata , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/terapia , Activador de Tejido Plasminógeno/uso terapéutico
19.
Pharmacol Res ; 125(Pt B): 232-245, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28855094

RESUMEN

The lipid mediator sphingosine 1-phosphate (S1P) affects cellular functions in most systems. Interest in its therapeutic potential has increased following the discovery of its G protein-coupled receptors and the recent availability of agents that can be safely administered in humans. Although the role of S1P in bone biology has been the focus of much less research than its role in the nervous, cardiovascular and immune systems, it is becoming clear that this lipid influences many of the functions, pathways and cell types that play a key role in bone maintenance and repair. Indeed, S1P is implicated in many osteogenesis-related processes including stem cell recruitment and subsequent differentiation, differentiation and survival of osteoblasts, and coupling of the latter cell type with osteoclasts. In addition, S1P's role in promoting angiogenesis is well-established. The pleiotropic effects of S1P on bone and blood vessels have significant potential therapeutic implications, as current therapeutic approaches for critical bone defects show significant limitations. Because of the complex effects of S1P on bone, the pharmacology of S1P-like agents and their physico-chemical properties, it is likely that therapeutic delivery of S1P agents will offer significant advantages compared to larger molecular weight factors. Hence, it is important to explore novel methods of utilizing S1P agents therapeutically, and improve our understanding of how S1P and its receptors modulate bone physiology and repair.


Asunto(s)
Huesos/metabolismo , Lisofosfolípidos/metabolismo , Osteogénesis/fisiología , Esfingosina/análogos & derivados , Animales , Humanos , Neovascularización Fisiológica/fisiología , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Transducción de Señal , Esfingosina/metabolismo , Células Madre/metabolismo
20.
Cell Death Dis ; 8(7): e2912, 2017 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-28682313

RESUMEN

Our previous findings suggest that sphingosine kinase 2 (SPK2) mediates ischemic tolerance and autophagy in cerebral preconditioning. The aim of this study was to determine by which mechanism SPK2 activates autophagy in neural cells. In both primary murine cortical neurons and HT22 hippocampal neuronal cells, overexpression of SPK2 increased LC3II and enhanced the autophagy flux. SPK2 overexpression protected cortical neurons against oxygen glucose deprivation (OGD) injury, as evidenced by improvement of neuronal morphology, increased cell viability and reduced lactate dehydrogenase release. The inhibition of autophagy effectively suppressed the neuroprotective effect of SPK2. SPK2 overexpression reduced the co-immunoprecipitation of Beclin-1 and Bcl-2, while Beclin-1 knockdown inhibited SPK2-induced autophagy. Both co-immunoprecipitation and GST pull-down analysis suggest that SPK2 directly interacts with Bcl-2. SPK2 might interact to Bcl-2 in the cytoplasm. Notably, an SPK2 mutant with L219A substitution in its putative BH3 domain was not able to activate autophagy. A Tat peptide fused to an 18-amino acid peptide encompassing the native, but not the L219A mutated BH3 domain of SPK2 activated autophagy in neural cells. The Tat-SPK2 peptide also protected neurons against OGD injury through autophagy activation. These results suggest that SPK2 interacts with Bcl-2 via its BH3 domain, thereby dissociating it from Beclin-1 and activating autophagy. The observation that Tat-SPK2 peptide designed from the BH3 domain of SPK2 activates autophagy and protects neural cells against OGD injury suggest that this structure may provide the basis for a novel class of therapeutic agents against ischemic stroke.


Asunto(s)
Isquemia Encefálica/genética , Neuronas/metabolismo , Fragmentos de Péptidos/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Recombinantes de Fusión/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Secuencia de Aminoácidos , Animales , Autofagia/genética , Beclina-1/genética , Beclina-1/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Regulación de la Expresión Génica , Glucosa/deficiencia , Hipocampo/metabolismo , Hipocampo/patología , Precondicionamiento Isquémico/métodos , Isoflurano/administración & dosificación , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/patología , Fragmentos de Péptidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Cultivo Primario de Células , Unión Proteica , Dominios Proteicos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...