Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 29(4): 1439-1458, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33309882

RESUMEN

Contrasting myelin damage through the generation of new myelinating oligodendrocytes represents a promising approach to promote functional recovery after stroke. Here, we asked whether activation of microglia and monocyte-derived macrophages affects the regenerative process sustained by G protein-coupled receptor 17 (GPR17)-expressing oligodendrocyte precursor cells (OPCs), a subpopulation of OPCs specifically reacting to ischemic injury. GPR17-iCreERT2:CAG-eGFP reporter mice were employed to trace the fate of GPR17-expressing OPCs, labeled by the green fluorescent protein (GFP), after permanent middle cerebral artery occlusion. By microglia/macrophages pharmacological depletion studies, we show that innate immune cells favor GFP+ OPC reaction and limit myelin damage early after injury, whereas they lose their pro-resolving capacity and acquire a dystrophic "senescent-like" phenotype at later stages. Intracerebral infusion of regenerative microglia-derived extracellular vesicles (EVs) restores protective microglia/macrophages functions, limiting their senescence during the post-stroke phase, and enhances the maturation of GFP+ OPCs at lesion borders, resulting in ameliorated neurological functionality. In vitro experiments show that EV-carried transmembrane tumor necrosis factor (tmTNF) mediates the pro-differentiating effects on OPCs, with future implications for regenerative therapies.


Asunto(s)
Senescencia Celular/genética , Vaina de Mielina/genética , Receptores Acoplados a Proteínas G/genética , Accidente Cerebrovascular/terapia , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Diferenciación Celular/genética , Línea Celular , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/terapia , Macrófagos/metabolismo , Macrófagos/trasplante , Masculino , Ratones , Microglía/metabolismo , Microglía/trasplante , Oligodendroglía/trasplante , Medicina Regenerativa/métodos , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología , Factor de Necrosis Tumoral alfa/genética
2.
Eur Heart J ; 42(32): 3078-3090, 2021 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-34252181

RESUMEN

AIMS: PCSK9 is secreted into the circulation, mainly by the liver, and interacts with low-density lipoprotein receptor (LDLR) homologous and non-homologous receptors, including CD36, thus favouring their intracellular degradation. As PCSK9 deficiency increases the expression of lipids and lipoprotein receptors, thus contributing to cellular lipid accumulation, we investigated whether this could affect heart metabolism and function. METHODS AND RESULTS: Wild-type (WT), Pcsk9 KO, Liver conditional Pcsk9 KO and Pcsk9/Ldlr double KO male mice were fed for 20 weeks with a standard fat diet and then exercise resistance, muscle strength, and heart characteristics were evaluated. Pcsk9 KO presented reduced running resistance coupled to echocardiographic abnormalities suggestive of heart failure with preserved ejection fraction (HFpEF). Heart mitochondrial activity, following maximal coupled and uncoupled respiration, was reduced in Pcsk9 KO mice compared to WT mice and was coupled to major changes in cardiac metabolism together with increased expression of LDLR and CD36 and with lipid accumulation. A similar phenotype was observed in Pcsk9/Ldlr DKO, thus excluding a contribution for LDLR to cardiac impairment observed in Pcsk9 KO mice. Heart function profiling of the liver selective Pcsk9 KO model further excluded the involvement of circulating PCSK9 in the development of HFpEF, pointing to a possible role locally produced PCSK9. Concordantly, carriers of the R46L loss-of-function variant for PCSK9 presented increased left ventricular mass but similar ejection fraction compared to matched control subjects. CONCLUSION: PCSK9 deficiency impacts cardiac lipid metabolism in an LDLR independent manner and contributes to the development of HFpEF.


Asunto(s)
Insuficiencia Cardíaca , Proproteína Convertasa 9 , Animales , Insuficiencia Cardíaca/genética , Masculino , Ratones , Ratones Noqueados , Proproteína Convertasa 9/genética , Receptores de LDL/genética , Volumen Sistólico
3.
Int J Mol Sci ; 22(22)2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34830207

RESUMEN

Nearly 18 million people died from cardiovascular diseases in 2019, of these 85% were due to heart attack and stroke. The available therapies although efficacious, have narrow therapeutic window and long list of contraindications. Therefore, there is still an urgent need to find novel molecular targets that could protect the brain and heart against ischemia without evoking major side effects. Nuclear receptors are one of the promising targets for anti-ischemic drugs. Modulation of estrogen receptors (ERs) and peroxisome proliferator-activated receptors (PPARs) by their ligands is known to exert neuro-, and cardioprotective effects through anti-apoptotic, anti-inflammatory or anti-oxidant action. Recently, it has been shown that the expression of aryl hydrocarbon receptor (AhR) is strongly increased after brain or heart ischemia and evokes an activation of apoptosis or inflammation in injury site. We hypothesize that activation of ERs and PPARs and inhibition of AhR signaling pathways could be a promising strategy to protect the heart and the brain against ischemia. In this Review, we will discuss currently available knowledge on the mechanisms of action of ERs, PPARs and AhR in experimental models of stroke and myocardial infarction and future perspectives to use them as novel targets in cardiovascular diseases.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Miocárdica/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Estrógenos/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Isquemia Encefálica/tratamiento farmacológico , Modelos Animales de Enfermedad , Humanos , Ligandos , Ratones , Terapia Molecular Dirigida/métodos , Isquemia Miocárdica/tratamiento farmacológico , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Resultado del Tratamiento
4.
Pharmacol Res ; 142: 223-236, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30818044

RESUMEN

Stroke is one of the main causes of death, neurological dysfunctions or disability in elderly. Neuroprotective drugs have been proposed to improve long-term recovery after stroke, but failed to reach clinical effectiveness. Hence, recent studies suggested that restorative therapies should combine neuroprotection and remyelination. Montelukast, an anti-asthmatic drug, was shown to exert neuroprotection in animal models of CNS injuries, but its ability to affect oligodendrocytes, restoring fiber connectivity, remains to be determined. In this study, we evaluated whether montelukast induces long-term repair by promoting fiber connectivity up to 8 weeks after middle cerebral artery occlusion (MCAo), using different experimental approaches such as in vivo diffusion magnetic resonance imaging (MRI), electrophysiological techniques, ex vivo diffusion tensor imaging (DTI)-based fiber tracking and immunohistochemistry. We found that, in parallel with a reduced evolution of ischemic lesion and atrophy, montelukast increased the DTI-derived axial diffusivity and number of myelin fibers, the density of myelin binding protein (MBP) and the number of GSTpi+ mature oligodendrocytes. Together with the rescue of MCAo-induced impairments of local field potentials in ischemic cortex, the data suggest that montelukast may improve fibers reorganization. Thus, to ascertain whether this effect involved changes of oligodendrocyte precursor cells (OPCs) activation and maturation, we used the reporter GPR17iCreERT2:CAG-eGreen florescent protein (GFP) mice that allowed us to trace the fate of OPCs throughout animal's life. Our results showed that montelukast enhanced the OPC recruitment and proliferation at acute phase, and increased their differentiation to mature oligodendrocytes at chronic phase after MCAo. Considering the crosstalk between OPCs and microglia has been widely reported in the context of demyelinating insults, we also assessed microglia activation. We observed that montelukast influenced the phenotype of microglial cells, increasing the number of M2 polarized microglia/macrophages, over the M1 phenotype, at acute phase after MCAo. In conclusion, we demonstrated that montelukast improves fiber re-organization and long-term functional recovery after brain ischemia, enhancing recruitment and maturation of OPCs. The present data suggest that montelukast, an already approved drug, could be "repositioned "as a protective drug in stroke acting also on fiber re-organization.


Asunto(s)
Acetatos/uso terapéutico , Antiasmáticos/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Quinolinas/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Encéfalo/patología , Ciclopropanos , Infarto de la Arteria Cerebral Media/fisiopatología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Microglía/efectos de los fármacos , Accidente Cerebrovascular/fisiopatología , Sulfuros
5.
Mediators Inflamm ; 2017: 3454212, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28607533

RESUMEN

Cysteinyl leukotrienes (CysLTs) are potent lipid mediators widely known for their actions in asthma and in allergic rhinitis. Accumulating data highlights their involvement in a broader range of inflammation-associated diseases such as cancer, atopic dermatitis, rheumatoid arthritis, and cardiovascular diseases. The reported elevated levels of CysLTs in acute and chronic brain lesions, the association between the genetic polymorphisms in the LTs biosynthesis pathways and the risk of cerebral pathological events, and the evidence from animal models link also CysLTs and brain diseases. This review will give an overview of how far research has gone into the evaluation of the role of CysLTs in the most prevalent neurodegenerative disorders (ischemia, Alzheimer's and Parkinson's diseases, multiple sclerosis/experimental autoimmune encephalomyelitis, and epilepsy) in order to understand the underlying mechanism by which they might be central in the disease progression.


Asunto(s)
Cisteína/metabolismo , Leucotrienos/metabolismo , Animales , Artritis Reumatoide/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedades del Sistema Nervioso Central/metabolismo , Dermatitis Atópica/metabolismo , Humanos , Enfermedades Neurodegenerativas/metabolismo
6.
Mediators Inflamm ; 2017: 2432958, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28932020

RESUMEN

Cysteinyl leukotrienes (CysLTs) are potent lipid inflammatory mediators synthesized from arachidonic acid, through the 5-lipoxygenase (5-LO) pathway. Owing to their properties, CysLTs play a crucial role in the pathogenesis of inflammation; therefore, CysLT modifiers as synthesis inhibitors or receptor antagonists, central in asthma management, may become a potential target for the treatment of other inflammatory diseases such as the cardiovascular disorders. 5-LO pathway activation and increased expression of its mediators and receptors are found in cardiovascular diseases. Moreover, the cardioprotective effects observed by using CysLT modifiers are promising and contribute to elucidate the link between CysLTs and cardiovascular disease. The aim of this review is to summarize the state of present research about the role of the CysLTs in the pathogenesis and progression of atherosclerosis and myocardial infarction.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Cisteína/metabolismo , Leucotrienos/metabolismo , Animales , Araquidonato 5-Lipooxigenasa/metabolismo , Aterosclerosis/metabolismo , Humanos , Infarto del Miocardio/metabolismo
7.
Glia ; 64(2): 287-99, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26464068

RESUMEN

In the adult brain NG2-glia continuously generate mature, myelinating oligodendrocytes. To which extent the differentiation process is common to all NG2-glia and whether distinct pools are recruited for repair under physiological and pathological conditions still needs clarification. Here, we aimed at investigating the differentiation potential of adult NG2-glia that specifically express the G-protein coupled receptor 17 (GPR17), a membrane receptor that regulates the differentiation of these cells at postnatal stages. To this aim, we generated the first BAC transgenic GPR17-iCreER(T2) mouse line for fate mapping studies. In these mice, under physiological conditions, GPR17(+) cells--in contrast to GPR17(-) NG2-glia--did not differentiate within 3 months, a peculiarity that was overcome after cerebral damage induced by acute injury or ischemia. After these insults, GPR17(+) NG2-glia rapidly reacted to the damage and underwent maturation, suggesting that they represent a 'reserve pool' of adult progenitors maintained for repair purposes.


Asunto(s)
Antígenos/metabolismo , Lesiones Encefálicas/fisiopatología , Isquemia Encefálica/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/fisiología , Oligodendroglía/fisiología , Proteoglicanos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Encéfalo/patología , Encéfalo/fisiología , Encéfalo/fisiopatología , Lesiones Encefálicas/patología , Isquemia Encefálica/patología , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Infarto de la Arteria Cerebral Media , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/patología , Neurogénesis/fisiología , Oligodendroglía/patología , Receptores Acoplados a Proteínas G/genética , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo
8.
J Cell Mol Med ; 18(9): 1785-96, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24909956

RESUMEN

GPR17 is a G(i) -coupled dual receptor activated by uracil-nucleotides and cysteinyl-leukotrienes. These mediators are massively released into hypoxic tissues. In the normal heart, GPR17 expression has been reported. By contrast, its role in myocardial ischaemia has not yet been assessed. In the present report, the expression of GPR17 was investigated in mice before and at early stages after myocardial infarction by using immunofluorescence, flow cytometry and RT-PCR. Before induction of ischaemia, results indicated the presence of the receptor in a population of stromal cells expressing the stem-cell antigen-1 (Sca-1). At early stages after ligation of the coronary artery, the receptor was expressed in Sca-1(+) cells, and cells stained with Isolectin-B4 and anti-CD45 antibody. GPR17(+) cells also expressed mesenchymal marker CD44. GPR17 function was investigated in vitro in a Sca-1(+)/CD31(-) cell line derived from normal hearts. These experiments showed a migratory function of the receptor by treatment with UDP-glucose and leukotriene LTD4, two GPR17 pharmacological agonists. The GPR17 function was finally assessed in vivo by treating infarcted mice with Cangrelor, a pharmacological receptor antagonist, which, at least in part, inhibited early recruitment of GPR17(+) and CD45(+) cells. These findings suggest a regulation of heart-resident mesenchymal cells and blood-borne cellular species recruitment following myocardial infarction, orchestrated by GPR17.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Infarto del Miocardio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Animales , Antígenos Ly/metabolismo , Movimiento Celular , Receptores de Hialuranos , Antígenos Comunes de Leucocito/metabolismo , Leucotrieno D4/farmacología , Leucotrieno D4/fisiología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Proteínas del Tejido Nervioso/agonistas , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Acoplados a Proteínas G/agonistas , Uridina Difosfato Glucosa/farmacología , Uridina Difosfato Glucosa/fisiología
9.
Sci Rep ; 14(1): 3371, 2024 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-38337010

RESUMEN

Preclinical and clinical data indicate that the 5-lipoxygenase pathway becomes activated in cardiovascular diseases suggesting an important role of CysLTs in atherosclerosis and in its ischemic complications. This study aims to investigate the effects of montelukast, a CysLTR-1 antagonist, in a mouse model of myocardial infarction (MI). C57BL/6N female mice were subjected to coronary artery ligation and received montelukast (10 mg/kg/day, intraperitoneal) or vehicle. Montelukast exerted beneficial effects in the infarcted area, decreasing mRNA expression of inflammatory genes, such Il1ß and Ccl2 (p < 0.05), at 48 h after MI, and reducing infarct size and preventing ischemic wall thinning (p < 0.05) at 4 weeks. Furthermore, montelukast counteracted maladaptive remodelling of whole heart. Indeed, montelukast reduced LV mass (p < 0.05) and remote wall thickening (p < 0.05), and improved cardiac pumping function, as evidenced by increased global ejection fraction (p < 0.01), and regional contractility in infarcted (p < 0.05) and in remote non-infarcted (p < 0.05) myocardium. Finally, montelukast prevented cardiomyocytes hypertrophy (p < 0.05) in remote myocardium, reducing the phosphorylation of GSK3ß, a regulator of hypertrophic pathway (p < 0.05). Our data strongly demonstrate the ability of montelukast to contrast the MI-induced maladaptive conditions, thus sustaining cardiac contractility. The results provide evidences for montelukast "repurposing" in cardiovascular diseases and in particular in myocardial infarction.


Asunto(s)
Acetatos , Ciclopropanos , Infarto del Miocardio , Quinolinas , Sulfuros , Remodelación Ventricular , Ratones , Animales , Femenino , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo , Miocardio/metabolismo
10.
Eur J Pharmacol ; 978: 176767, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-38909934

RESUMEN

Fenofibrate, a PPAR-α agonist clinically used to lower serum lipid levels, reduces cardiac remodeling and improves cardiac function. However, its mechanism of action is not completely elucidated. In this study we examined the effect of fenofibrate on mitochondria in a rat model of renovascular hypertension, focusing on mediators controlling mitochondrial dynamics and autophagy. Rats with two-kidney one-clip (2K1C) hypertension were treated with fenofibrate 150 mg/kg/day (2K1C-FFB) or vehicle (2K1C-VEH) for 8 weeks. Systolic blood pressure and cardiac functional were in-vivo assessed, while cardiomyocyte size and protein expression of mediators of cardiac hypertrophy and mitochondrial dynamics were ex-vivo examined by histological and Western blot analyses. Fenofibrate treatment counteracted the development of hypertension and the increase of left ventricular mass, relative wall thickness and cross-sectional area of cardiomyocytes. Furthermore, fenofibrate re-balanced the expression Mfn2, Drp1 and Parkin, regulators of fusion, fission, mitophagy respectively. Regarding autophagy, the LC3-II/LC3-I ratio was increased in 2K1C-VEH and 2K1C-FFB, whereas the autophagy was increased only in 2K1C-FFB. In cultured H9C2 cardiomyoblasts, fenofibrate reversed the Ang II-induced mRNA up-regulation of hypertrophy markers Nppa and Myh7, accumulation of reactive oxygen species and depolarization of the mitochondrial membrane exerting protection mediated by up-regulation of the Uncoupling protein 2. Our results indicate that fenofibrate acts directly on cardiomyocytes and counteracts the pressure overload-induced cardiac maladaptive remodeling. This study reveals a so far hidden mechanism involving mitochondrial dynamics in the beneficial effects of fenofibrate, support its repurposing for the treatment of cardiac hypertrophy and provide new potential targets for its pharmacological function.


Asunto(s)
Cardiomegalia , Modelos Animales de Enfermedad , Fenofibrato , Dinámicas Mitocondriales , Miocitos Cardíacos , Remodelación Ventricular , Animales , Fenofibrato/farmacología , Fenofibrato/uso terapéutico , Dinámicas Mitocondriales/efectos de los fármacos , Masculino , Ratas , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/metabolismo , Cardiomegalia/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Remodelación Ventricular/efectos de los fármacos , Autofagia/efectos de los fármacos , Hipertensión Renovascular/tratamiento farmacológico , Hipertensión Renovascular/metabolismo , Hipertensión Renovascular/patología , Hipertensión Renovascular/fisiopatología , Ratas Wistar , Presión Sanguínea/efectos de los fármacos
11.
Biomed Pharmacother ; 172: 116201, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38306846

RESUMEN

The treatment of glioblastoma (GBM) faces significant challenges due to the difficulty of delivering drugs through the blood-brain barrier (BBB). Extracellular vesicles (EVs) have emerged as potential carriers for targeted drug delivery to brain tumors. However, their use and distribution in the presence of an intact BBB and their ability to target GBM tissue are still under investigation. This study explored the use of EVs for GBM targeting across the BBB. Canine plasma EVs from healthy dogs and dogs with glioma were isolated, characterized, and loaded with diagnostic agents. Biodistribution studies were conducted in healthy murine models and a novel intranasal model that preserved BBB integrity while initiating early-stage GBM growth. This model assessed EVs' potential for delivering the contrast agent gadoteric acid to intracranial tumors. Imaging techniques, such as bioluminescence and MRI, confirmed EVs' targeting and delivery capabilities thus revealing a selective accumulation of canine glioma-derived EVs in brain tissue under physiological conditions. In the model of brain tumor, MRI experiments demonstrated the ability of EVs to accumulate gadoteric acid within GBM to enhance contrast of the tumoral mass, even when BBB integrity is maintained. This study underscores the potential of EVs derived from glioma for the targeted delivery of drugs to glioblastoma. EVs from dogs with glioma showed capacity to traverse the BBB and selectively accumulate within the brain tumor. Overall, this research represents a foundation for the application of autologous EVs to precision glioblastoma treatment, addressing the challenge of BBB penetration and targeting specificity in brain cancer therapy.


Asunto(s)
Neoplasias Encefálicas , Vesículas Extracelulares , Glioblastoma , Glioma , Perros , Animales , Ratones , Glioblastoma/diagnóstico por imagen , Barrera Hematoencefálica , Distribución Tisular , Neoplasias Encefálicas/diagnóstico por imagen , Quelantes , Medios de Contraste
12.
Neuroscience ; 523: 1-6, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37211082

RESUMEN

Cerebral collaterals are recruited after arterial occlusion with a protective effect on tissue outcome in acute ischemic stroke. Head down tilt 15° (HDT15) is a simple, low cost and accessible procedure that could be applied as an emergency treatment, before recanalization therapies, with the aim to increase cerebral collateral flow. Spontaneously hypertensive rats have been shown to display anatomical differences in morphology and function of cerebral collaterals, compared to other rat strains, resulting in an overall poor collateral circulation. We investigate the efficacy and safety of HDT15 in spontaneously hypertensive (SHR) rats, which were considered as an animal stroke model with poor collaterals. Cerebral ischemia was induced by 90 minute endovascular occlusion of the middle cerebral artery (MCA). SHR rats were randomized to HDT15 or flat position (n = 19). HDT15 was applied 30 minutes after occlusion and lasted 60 minutes, until reperfusion. HDT15 application increased cerebral perfusion (+16.6% versus +6.1%; p = 0.0040) and resulted in a small reduction of infarct size (83.6 versus 107.1 mm3; - 21.89%; p = 0.0272), but it was not associated with early neurological improvement, compared to flat position. Our study suggests that the response to HDT15 during MCA occlusion is dependent on baseline collaterals. Nonetheless, HDT15 promoted a mild improvement of cerebral hemodynamics even in subjects with poor collaterals, without safety concerns.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Animales , Ratas , Circulación Cerebrovascular/fisiología , Inclinación de Cabeza , Ratas Endogámicas SHR
13.
J Cereb Blood Flow Metab ; 43(7): 1077-1088, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36823998

RESUMEN

Multicentre preclinical randomized controlled trials (pRCTs) are a valuable tool to improve experimental stroke research, but are challenging and therefore underused. A common challenge regards the standardization of procedures across centres. We here present the harmonization phase for the quantification of sensorimotor deficits by composite neuroscore, which was the primary outcome of two multicentre pRCTs assessing remote ischemic conditioning in rodent models of ischemic stroke. Ischemic stroke was induced by middle cerebral artery occlusion for 30, 45 or 60 min in mice and 50, 75 or 100 min in rats, allowing sufficient variability. Eleven animals per species were video recorded during neurobehavioural tasks and evaluated with neuroscore by eight independent raters, remotely and blindly. We aimed at reaching an intraclass correlation coefficient (ICC) ≥0.60 as satisfactory interrater agreement. After a first remote training we obtained ICC = 0.50 for mice and ICC = 0.49 for rats. Errors were identified in animal handling and test execution. After a second remote training, we reached the target interrater agreement for mice (ICC = 0.64) and rats (ICC = 0.69). In conclusion, a multi-step, online harmonization phase proved to be feasible, easy to implement and highly effective to align each centre's behavioral evaluations before project's interventional phase.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Ratas , Ratones , Animales , Infarto de la Arteria Cerebral Media , Ensayos Clínicos Controlados Aleatorios como Asunto
14.
J Voice ; 36(2): 288.e1-288.e14, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32768157

RESUMEN

OBJECTIVES: The esophageal speech is one of the possible alaryngeal voices resulting after total laryngectomy. Its production is made by the regurgitation of the air coming from the esophagus, sonorized through the passage from the walls of the upper esophageal sphincter. The neural correlates of this voice have never been investigated, while the neural control of laryngeal voice has been already documented by different studies. METHODS: Four patients using esophageal speech after total laryngectomy and four healthy controls underwent functional magnetic resonance imaging. The fMRI experiment was carried out using a "Block Design Paradigm." RESULTS: Comparison of the phonation task in the two groups revealed higher brain activities in the cingulate gyrus, the cerebellum and the medulla as well as lower brain activities in the precentral gyrus, the inferior and middle frontal gyrus and the superior temporal gyrus in the laryngectomized group. CONCLUSIONS: The findings in this pilot study provide insight into neural phonation control in laryngectomized patients with esophageal speech. The imaging results demonstrated that in patients with esophageal speech, altered brain activities can be observed. The adaptive changes in the brain following laryngectomy reflect the changes in the body and in the voice modality. In addition, this pilot study establishes that a blocked design fMRI is sensitive enough to define a neural network associated with esophageal voice and lays the foundation for further studies in this field.


Asunto(s)
Laringe Artificial , Voz Alaríngea , Humanos , Laringectomía/efectos adversos , Imagen por Resonancia Magnética , Fonación , Proyectos Piloto , Voz Esofágica
15.
J Mol Med (Berl) ; 100(1): 23-41, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34674004

RESUMEN

Myocardial infarction (MI) is the leading cause of death among ischemic heart diseases and is associated with several long-term cardiovascular complications, such as angina, re-infarction, arrhythmias, and heart failure. However, MI is frequently accompanied by non-cardiovascular multiple comorbidities, including brain disorders such as stroke, anxiety, depression, and cognitive impairment. Accumulating experimental and clinical evidence suggests a causal relationship between MI and stroke, but the precise underlying mechanisms have not yet been elucidated. Indeed, the risk of stroke remains a current challenge in patients with MI, in spite of the improvement of medical treatment among this patient population has reduced the risk of stroke. In this review, the effects of the signaling from the ischemic heart to the brain, such as neuroinflammation, neuronal apoptosis, and neurogenesis, and the possible actors mediating these effects, such as systemic inflammation, immunoresponse, extracellular vesicles, and microRNAs, are discussed.


Asunto(s)
Encéfalo , Infarto del Miocardio , Miocardio , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Humanos , Infarto del Miocardio/genética , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Miocardio/inmunología , Miocardio/metabolismo , Transducción de Señal
16.
Am J Physiol Heart Circ Physiol ; 300(3): H762-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21148758

RESUMEN

Thromboxane A(2) and other eicosanoids such as isoprostanes contribute to vascular proliferation and atherosclerosis by binding to the thromboxane/prostaglandin endoperoxide receptors. The effects of terutroban, a thromboxane/prostaglandin endoperoxide receptor antagonist, on aorta remodeling were evaluated in spontaneously hypertensive stroke-prone rats (SHRSPs), a model of severe hypertension, endothelial dysfunction, vascular inflammation, and cerebrovascular diseases. Male SHRSPs were allocated to three groups receiving a standard diet (n = 5) or a high-sodium permissive diet plus vehicle (n = 6) or plus terutroban (30 mg · kg(-1) · day(-1); n = 6). After 6 wk of dietary treatment, all of the animals were injected with bromodeoxyuridine and simultaneously euthanized for aorta collection. The aortic media thickness-to-lumen ratio significantly (P < 0.0001) increased in the salt-loaded rats compared with the rats fed a standard diet, whereas terutroban treatment completely prevented media thickening (P < 0.001). When compared with vehicle, terutroban was also effective in preventing cell proliferation in the media, as indicated by the reduced number of bromodeoxyuridine-positive (P < 0.0001) and proliferating cell nuclear antigen-positive cells (P < 0.0001). Severe fibrosis characterized by a significant accumulation of collagen and fibronectin in the vascular wall was observed in the vehicle-treated rats (P < 0.01) but was completely prevented by terutroban (P < 0.001). The latter also inhibited heat shock protein-47 (P < 0.01) and TGF-1ß expression (P < 0.001), which were significantly increased by the high-salt diet. In conclusion, terutroban prevents the development of aorta hyperplasia and has beneficial effects on fibrotic processes by affecting TGF-ß and heat shock protein-47 expression in SHRSPs. These findings provide mechanistic data supporting the beneficial effects of terutroban in preventing or retarding atherogenesis.


Asunto(s)
Aorta/efectos de los fármacos , Aterosclerosis/prevención & control , Endotelio Vascular/efectos de los fármacos , Hiperplasia/prevención & control , Naftalenos/uso terapéutico , Propionatos/uso terapéutico , Receptores de Prostaglandina/antagonistas & inhibidores , Túnica Media/efectos de los fármacos , Animales , Aorta/química , Aorta/patología , Aterosclerosis/tratamiento farmacológico , Colágeno/metabolismo , Endotelio Vascular/metabolismo , Fibronectinas/metabolismo , Fibrosis , Hiperplasia/tratamiento farmacológico , Hipertrofia/patología , Masculino , Ratas , Ratas Endogámicas SHR , Cloruro de Sodio Dietético/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Túnica Media/metabolismo , Túnica Media/patología
17.
J Colloid Interface Sci ; 582(Pt B): 678-700, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-32911414

RESUMEN

HYPOTHESIS: Iron oxide and other ferrite nanoparticles have not yet found widespread application in the medical field since the translation process faces several big hurdles. The incomplete knowledge of the interactions between nanoparticles and living organisms is an unfavorable factor. This complex subject should be made simpler by synthesizing magnetic nanoparticles with good physical (relaxivity) and chemical (colloidal stability, anti-fouling) properties and no biological activity (no immune-related effects, minimal internalization, fast clearance). Such an innocent scaffold is the main aim of the present paper. We systematically searched for it within the class of small-to-medium size ferrite nanoparticles coated by small (zwitter)ionic ligands. Once established, it can be functionalized to achieve targeting, drug delivery, etc. and the observed biological effects will be traced back to the functional molecules only, as the nanosized scaffold is innocent. EXPERIMENTS: We synthesized nine types of magnetic nanoparticles by systematic variation of core composition, size, coating. We investigated their physico-chemical properties and interaction with serum proteins, phagocytic microglial cells, and a human model of inflammation and studied their biodistribution and clearance in healthy mice. The nanoparticles have good magnetic properties and their surface charge is determined by the preferential adsorption of anions. All nanoparticle types can be considered as immunologically safe, an indispensable pre-requisite for medical applications in humans. All but one type display low internalization by microglial BV2 cells, a process strongly affected by the nanoparticle size. Both small (3 nm) and medium size (11 nm) zwitterionic nanoparticles are in part captured by the mononuclear phagocyte system (liver and spleen) and in part rapidly (≈1 h) excreted through the urinary system of mice. FINDINGS: The latter result questions the universality of the accepted size threshold for the renal clearance of nanoparticles (5.5 nm). We suggest that it depends on the nature of the circulating particles. Renal filterability of medium-size magnetic nanoparticles is appealing because they share with small nanoparticles the decreased accumulation-related toxicity while performing better as magnetic diagnostic/therapeutic agents thanks to their larger magnetic moment. In conclusion, many of our nanoparticle types are a bio-compatible innocent scaffold with unexpectedly favorable clearance.


Asunto(s)
Nanopartículas de Magnetita , Nanopartículas , Animales , Proteínas Sanguíneas , Compuestos Férricos , Ratones , Distribución Tisular
18.
J Pharmacol Exp Ther ; 335(2): 324-31, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20671072

RESUMEN

A growing body of evidence suggests that chronic kidney disease is a significant risk for cardiovascular events and stroke regardless of traditional risk factors. The aim of this study was to examine the effects of peroxisome proliferator-activated receptor (PPAR) agonists on the tissue damage affecting salt-loaded spontaneously hypertensive stroke-prone rats ( SHRSPs), an animal model that develops a complex pathology characterized by systemic inflammation, hypertension, and proteinuria and leads to end-organ injury (initially renal and subsequently cerebral). Compared with the PPARγ agonist rosiglitazone, the PPARα ligands fenofibrate and clofibrate significantly increased survival (p < 0.001) by delaying the occurrence of brain lesions monitored by magnetic resonance imaging (p < 0.001) and delaying increased proteinuria (p < 0.001). Fenofibrate completely prevented the renal disorder characterized by severe vascular lesions, tubular damage, and glomerular sclerosis, reduced the number of ED-1-positive cells and collagen accumulation, and decreased the renal expression of interleukin-1ß, transforming growth factor ß, and monocyte chemoattractant protein 1. It also prevented the plasma and urine accumulation of acute-phase and oxidized proteins, suggesting that the protection induced by PPARα agonists was at least partially caused by their anti-inflammatory and antioxidative properties. The results of this study demonstrate that PPAR agonism has beneficial effects on spontaneous brain and renal damage in SHRSPs by inhibiting systemic inflammation and oxidative stress, and they support carrying out future studies aimed at evaluating the effect of PPARα agonists on proteinuria and clinical outcomes in hypertensive patients with renal disease at increased risk of stroke.


Asunto(s)
Encéfalo/patología , Inflamación/prevención & control , Enfermedades Renales/prevención & control , Estrés Oxidativo/efectos de los fármacos , PPAR alfa/agonistas , Accidente Cerebrovascular/complicaciones , Animales , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Quimiocina CCL2/biosíntesis , Clofibrato/farmacología , Clofibrato/uso terapéutico , Modelos Animales de Enfermedad , Fenofibrato/farmacología , Fenofibrato/uso terapéutico , Hipertensión/complicaciones , Hipertensión/tratamiento farmacológico , Hipertensión/metabolismo , Hipertensión/patología , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/biosíntesis , Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Ligandos , Masculino , Ratas , Ratas Endogámicas SHR , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Factor de Crecimiento Transformador beta/biosíntesis
19.
J Pharmacol Exp Ther ; 334(1): 199-205, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20332187

RESUMEN

This study investigated the efficacy of terutroban, a specific thromboxane/prostaglandin endoperoxide receptor antagonist, on stroke incidence in spontaneously hypertensive stroke-prone rats (SHRSP). The effects of terutroban were compared with those of aspirin, another antiplatelet agent, and rosuvastatin, known to exert end-organ protection in SHRSP. Salt-loaded male SHRSP were treated orally once a day with vehicle, terutroban (30 mg/kg/day), aspirin (60 mg/kg/day), or rosuvastatin (10 mg/kg/day). Compared with vehicle, and regardless of any effect on blood pressure or serum thromboxane B(2) levels, terutroban significantly increased survival (p < 0.001) as a consequence of a delayed brain lesion occurrence monitored by magnetic resonance imaging (p < 0.001), and a delayed increase of proteinuria (p < 0.001). Terutroban decreased cerebral mRNA transcription of interleukin-1beta, transforming growth factor-beta, and monocyte chemoattractant protein-1 after 6 weeks of dietary treatment. Terutroban also prevented the accumulation of urinary acute-phase proteins at high molecular weight, identified as markers of systemic inflammation, and assessed longitudinally by one-dimensional electrophoresis. Terutroban also has protective effects on the vasculature as suggested by the preservation of endothelial function and endothelial nitric-oxide synthase expression in isolated carotid arteries. These effects are similar to those obtained with rosuvastatin, and superior to those of aspirin. Terutroban increases survival in SHRSP by reducing systemic inflammation as well as preserving endothelial function. These data support clinical development of terutroban in the prevention of cerebrovascular and cardiovascular complications of atherothrombosis.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Hipertensión/tratamiento farmacológico , Naftalenos/uso terapéutico , Propionatos/uso terapéutico , Sustancias Protectoras/uso terapéutico , Receptores de Prostaglandina/antagonistas & inhibidores , Receptores de Tromboxanos/antagonistas & inhibidores , Accidente Cerebrovascular/prevención & control , Síndrome de Respuesta Inflamatoria Sistémica/prevención & control , Animales , Aspirina/administración & dosificación , Aspirina/farmacología , Aspirina/uso terapéutico , Biomarcadores/análisis , Biomarcadores/sangre , Biomarcadores/orina , Presión Sanguínea/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Fluorobencenos/administración & dosificación , Fluorobencenos/farmacología , Fluorobencenos/uso terapéutico , Hipertensión/complicaciones , Hipertensión/inmunología , Hipertensión/metabolismo , Hipertensión/patología , Imagen por Resonancia Magnética , Masculino , Naftalenos/administración & dosificación , Naftalenos/farmacología , Propionatos/administración & dosificación , Propionatos/farmacología , Sustancias Protectoras/administración & dosificación , Sustancias Protectoras/farmacología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Ratas , Ratas Endogámicas SHR , Rosuvastatina Cálcica , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Análisis de Supervivencia , Síndrome de Respuesta Inflamatoria Sistémica/etiología , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo
20.
Biochem Pharmacol ; 177: 113895, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32145263

RESUMEN

Drug repurposing is a promising way in drug discovery to identify new therapeutic uses -different from the original medical indication- for existing drugs. It has many advantages over traditional approaches to de novo drug discovery, since it can significantly reduce healthcare costs and development timeline. In this review, we discuss the possible repurposing of drugs approved for cardiovascular diseases, such as ß-blockers, angiotensin converting enzyme inhibitors (ACE-Is), angiotensin II receptor blockers (ARBs), statins, aspirin, cardiac glycosides and low-molecular-weight heparins (LMWHs). Indeed, numerous experimental and epidemiological studies have reported promising anti-cancer activities for these drugs. It is worth mentioning, however, that the results of these studies are often controversial and very few data were obtained by controlled prospective clinical trials. Therefore, no final conclusion has yet been reached in this area and no final recommendations can be made. Moreover, ß-blockers, ARBs and statins showed promising results in randomised controlled trials (RCTs) where pathological conditions other than cancer were considered. The results obtained have led or may lead to new indications for these drugs. For each drug or class of drugs, the potential molecular mechanisms of action justifying repurposing, results obtained in vitro and in animal models and data from epidemiological and randomized studies are described.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Infecciones Bacterianas/tratamiento farmacológico , Enfermedades Cardiovasculares/tratamiento farmacológico , Reposicionamiento de Medicamentos/métodos , Síndrome de Marfan/tratamiento farmacológico , Trastornos Migrañosos/tratamiento farmacológico , Micosis/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Periodontitis/tratamiento farmacológico , Antagonistas Adrenérgicos beta/uso terapéutico , Antagonistas de Receptores de Angiotensina/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Aspirina/uso terapéutico , Glicósidos Cardíacos/uso terapéutico , Heparina de Bajo-Peso-Molecular/uso terapéutico , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA