Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Physiol ; 236(3): 1967-1979, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32730642

RESUMEN

The transplantation of mesenchymal stem cells (MSCs) is of main approaches in regenerative therapy for stroke. Due to the potential tumorigenicity and low survival rate of transplanted cells, focuses have been shifted from cell replacement to their paracrine effects. Therefore, stem cell-conditioned medium (CM) therapy has emerged as an alternative candidate. Here, we investigated the effect of CM derived from human embryonic MSCs on experimental ischemic stroke. Wistar rats underwent ischemic stroke by the right middle cerebral artery occlusion (MCAO). CM was infused either one time (1 hr post-MCAO) or three times (1, 24, and 48 hr post-MCAO) through guide cannula into the left lateral ventricle. Neurological functions were evaluated using Bederson's test and modified Neurological Severity Score on Days 1, 3, and 7 following MCAO. Infarction volumes and cerebral edema were measured on Days 3 and 7. growth-associated protein-43, synaptophysin, cAMP response element-binding protein, and phosphorylated-cAMP response element-binding protein levels were also assessed in peri-ischemic cortical tissue on Day 7 postsurgery. Our results indicated that three times injections of CM could significantly reduce body weight loss, mortality rate, infarct volumes, cerebral edema, and improve neurological deficits in MCAO rats. Moreover, three injections of CM could restore decreased levels of synaptic markers in MCAO rats up to its normal levels observed in the sham group. Our data suggest that using the CM obtained from embryonic stem cells-MSCs could be a potent therapeutic approach to attenuate cerebral ischemia insults which may be partly mediated through modulation of synaptic plasticity.


Asunto(s)
Encéfalo/patología , Medios de Cultivo Condicionados/farmacología , Células Madre Embrionarias Humanas/citología , Células Madre Mesenquimatosas/metabolismo , Accidente Cerebrovascular/patología , Sinapsis/patología , Animales , Biomarcadores/metabolismo , Peso Corporal/efectos de los fármacos , Infarto Encefálico/complicaciones , Infarto Encefálico/patología , Línea Celular , Edema/complicaciones , Edema/patología , Humanos , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Inyecciones Intraventriculares , Masculino , Neurogénesis/efectos de los fármacos , Ratas Wistar , Sinapsis/efectos de los fármacos
2.
Neurobiol Dis ; 156: 105399, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34029695

RESUMEN

Immune system hypersensitivity is believed to contribute to mental frailty in the elderly. Solid evidence indicates NOD-like receptor pyrin domain containing-3 (NLRP3)-inflammasome activation intimately connects aging-associated chronic inflammation (inflammaging) to senile cognitive decline. Thioredoxin interacting protein (TXNIP), an inducible protein involved in oxidative stress, is essential for NLRP3 inflammasome activity. This study aims to find whether TXNIP/NLRP3 inflammasome pathway is involved in senile dementia. According to our studies on sex-matched mice, TXNIP was significantly upregulated in aged animals, paralleled by the NLRP3-inflammasome over-activity leading to enhanced caspase-1 cleavage and IL-1ß maturation, in both sexes. This was closely associated with depletion of the anti-aging and cognition enhancing protein klotho, in aged males. Txnip knockout reversed age-related NLRP3-hyperactivity and enhanced thioredoxin (TRX) levels. Further, TXNIP inhibition along with verapamil replicated TXNIP/NLRP3-inflammasome downregulation in aged animals, with FOXO-1 and mTOR upregulation. These alterations concurred with substantial improvements in both cognitive and sensorimotor abilities. Together, these findings substantiate the pivotal role of TXNIP to drive inflammaging in parallel with klotho depletion and functional decline, and delineate thioredoxin system as a potential target to decelerate senile dementia.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Proteínas Portadoras/biosíntesis , Mediadores de Inflamación/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/biosíntesis , Tiorredoxinas/biosíntesis , Envejecimiento/genética , Envejecimiento/patología , Animales , Encéfalo/patología , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Femenino , Mediadores de Inflamación/antagonistas & inhibidores , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Estrés Oxidativo/fisiología , Tiorredoxinas/antagonistas & inhibidores , Tiorredoxinas/genética
3.
Nutr Neurosci ; 22(4): 264-272, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28946820

RESUMEN

OBJECTIVES: Coenzyme Q10 (CoQ10, ubiquinone) stands among the safest supplements in the elderly to protect against cardiovascular disorders. Noteworthy, CoQ10 deficiency is common in many surviving stroke patients as they are mostly prescribed statins for the secondary prevention of stroke incidence lifelong. Accordingly, the current study aims to experimentally examine whether CoQ10 supplementation in animals receiving atorvastatin may affect acute stroke-induced injury. METHODS: Adult rats underwent transient middle cerebral artery occlusion after atorvastatin pretreatment (5 or 10 mg/ kg/day; po; 30 days) with or without CoQ10 (200 mg/kg/day). After 24 hours ischemic/reperfusion injury, animals were subjected to functional assessments followed by cerebral molecular and histological to detect inflammation, apoptosis and oxidative stress. RESULTS: Animals dosed with 10 mg/kg presented the worst neurological function and brain damage in the acute phase of stroke injury. CoQ10 supplementation efficiently improved functional deficit and cerebral infarction in all stroke animals, particularly those exhibiting statin toxicity. Such benefits were associated with remarkable anti-inflammatory and anti-apoptotic effects, based on the analyzed tumor necrosis factor-α, interleukin-6, Bax/Bcl2 and cleaved caspase 3/9 immunoblots. Importantly, our fluoro-jade staining data indicated CoQ10 may revert the stroke-induced neurodegeneration. No parallel alteration was detected in stroke-induced oxidative stress as determined by malondialdehyde and 8-oxo-2'-deoxyguanosine levels. DISCUSSION: These data suggest that all stroke animals may benefit from CoQ10 administration through modulating inflammatory and degenerative pathways. This study provides empirical evidence for potential advantages of CoQ10 supplementation in atorvastatin-receiving patients which may not shadow its antioxidant properties.


Asunto(s)
Atorvastatina/administración & dosificación , Isquemia Encefálica/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Accidente Cerebrovascular/prevención & control , Ubiquinona/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Isquemia Encefálica/complicaciones , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Encefalitis/etiología , Encefalitis/prevención & control , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Resultado del Tratamiento , Ubiquinona/administración & dosificación
4.
Brain Inj ; 31(13-14): 1910-1917, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28898133

RESUMEN

OBJECTIVE: Physical exercise contributes to improving stability against nerve injury caused by ischaemic stroke. Here we aimed to preliminarily investigate the effects of continuous endurance training (CET) and high-intensity interval training (HIT) on stroke-associated anxiety, locomotion, neurological assessments and P70S6 Kinase (P70S6K) activation as well. To do this, rats were trained according to HIT and CET protocols for 2 months prior to being subject to middle cerebral artery occlusion surgery. METHODS: Twenty-four hours later behavioural examination was performed by elevated plus maze (EPM) testing, open field and neurological scoring followed by cortical and hippocampal P70S6Ks immunoblotting. RESULTS: According to the obtained data pre-ischaemic HIT and CET similarly improved neurological performance, anxiety levels and locomotion in EPM and open field tests following ischaemic stroke while there was a remarkable rise in hippocampal and cortical P70S6K activation in the HIT group compared to the CET counterparts. CONCLUSION: Behavioral and molecular data suggest that interval training is more beneficial rather than CET, but the distinct mechanisms of CET and HIT on memory are still topics to be discovered.


Asunto(s)
Infarto de la Arteria Cerebral Media/complicaciones , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/rehabilitación , Condicionamiento Físico Animal , Análisis de Varianza , Animales , Ansiedad/etiología , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Prueba de Esfuerzo , Conducta Exploratoria/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Infarto de la Arteria Cerebral Media/patología , Masculino , Aprendizaje por Laberinto/fisiología , Consumo de Oxígeno/fisiología , Fosforilación , Ratas , Reperfusión/efectos adversos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo
5.
Nutr Neurosci ; 24(7): 581-582, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-31455182
6.
Can J Neurol Sci ; 42(3): 195-202, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25860537

RESUMEN

BACKGROUND: Recent evidence suggests that an extreme shift may occur in sphingosine metabolism in neuroinflammatory contexts. Sphingosine 1-phosphate (S1P)-metabolizing enzymes (SMEs) regulate the level of S1P. We recently found that FTY720, a S1P analogue, and SEW2871, a selective S1P receptor 1 (S1P1) agonist, provide protection against neural damage and memory deficit in amyloid beta (Aß)-injected animals. This study aimed to evaluate the effects of these two analogues on the expression of SMEs as well as their anti-inflammatory roles. METHODS: Rats were treated with intracerebral lipopolysaccharide (LPS) or Aß. Memory impairment was assessed by Morris water maze and the effects of drugs on SMEs as well as inflammatory markers, TNF- α and COX-II, were determined by immunoblotting. RESULTS: Aß and LPS differentially altered the expression profile of SMEs. In Aß-injected animals, FTY720 and SEW2871 treatments exerted anti-inflammatory effects and restored the expression profile of SMEs, in parallel to our previous findings. In LPS animals however, in spite of anti-inflammatory effects of the two analogues, only FTY720 restored the levels of SMEs and prevented memory deficit. CONCLUSION: The observed ameliorating effects of FTY720 and SEW7821 can be partly attributed to the interruption of the vicious cycle of abnormal S1P metabolism and neuro-inflammation. The close imitation of the FTY720 effects by SW2871 in Aß-induced neuro-inflammation may highlight the attractive role of S1P1 as a potential target to restore S1P metabolism and inhibit inflammatory processes.


Asunto(s)
Inflamación/tratamiento farmacológico , Lisofosfolípidos/metabolismo , Receptores de Lisoesfingolípidos/agonistas , Esfingosina/análogos & derivados , Péptidos beta-Amiloides/administración & dosificación , Péptidos beta-Amiloides/metabolismo , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Ciclooxigenasa 2/metabolismo , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Inflamación/inducido químicamente , Inflamación/metabolismo , Lipopolisacáridos/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/psicología , Oxadiazoles/farmacología , Oxadiazoles/uso terapéutico , Ratas , Ratas Wistar , Esfingosina/metabolismo , Tiofenos/farmacología , Tiofenos/uso terapéutico , Factor de Necrosis Tumoral alfa/metabolismo
7.
Exp Brain Res ; 232(11): 3687-96, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25098558

RESUMEN

Lipopolysaccharide is an endotoxin to induce sickness behavior in several animal models to explore the link between immune activation and cognition. Neuroinflammation playing a pivotal role in disease progress is evidently influenced by sphingosine-1-phosphate. As one of the sphingosine analogs in clinical use for multiple sclerosis, fingolimod (FTY720) was shown to substantially affect gene expression profile in the context of AD in our previous experiments. The present study was designed to evaluate the drug efficacy in the context of the mere inflammatory context leading to memory impairment. FTY720 was repeatedly administered for a few days before or after intracerebral lipopolysaccharide (LPS) injection in rats. Animal's brains were then assigned to histological as well as multiplex mRNA assay following memory performance test. Both FTY720 pre-treatment and post-treatment were similarly capable of ameliorating LPS-induced memory impairment as assessed by passive avoidance test. Such amending effects may be partly accountable by the concomitant alterations in transcriptional levels of mitogen-activated protein kinases as well as inflammatory genes determined by QuantiGene Plex analysis. These findings confirming FTY720 application benefits suggest its efficacy may not differ significantly while considered either as a preventive or as a therapeutic approach against neuroinflammation.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/toxicidad , Trastornos de la Memoria/inducido químicamente , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Animales , Reacción de Prevención/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Caspasa 3/metabolismo , Adaptación a la Oscuridad/efectos de los fármacos , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Esquema de Medicación , Femenino , Clorhidrato de Fingolimod , Inmunosupresores/farmacología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/fisiopatología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Wistar , Esfingosina/farmacología , Transcriptoma , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
8.
Front Pharmacol ; 14: 1161999, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37124219

RESUMEN

The abrupt hyperglycemic reperfusion following thrombectomy has been shown to harm the efficacy of the intervention in stroke patients with large vessel occlusion. Studies of ours and others have shown thioredoxin-interacting protein (TXNIP) is critically involved in hyperglycemic stroke injury. We recently found verapamil ameliorates cerebrovascular toxicity of tissue plasminogen activators in hyperglycemic stroke. The present study aims to answer if verapamil exerts direct neuroprotective effects and alleviates glucose toxicity following thrombectomy in a preclinical model of hyperglycemic stroke. Primary cortical neural (PCN) cultures were exposed to hyperglycemic reperfusion following oxygen-glucose deprivation (OGD), with or without verapamil treatment. In a mouse model of intraluminal stroke, animals were subjected to 4 h middle cerebral artery occlusion (MCAO) and intravenous glucose infusion. Glucose infusion lasted one more hour at reperfusion, along with intra-arterial (i.a.) verapamil infusion. Animals were subjected to sensorimotor function tests and histological analysis of microglial phenotype at 72 h post-stroke. According to our findings, glucose concentrations (2.5-20 mM) directly correlated with TXNIP expression in OGD-exposed PCN cultures. Verapamil (100 nM) effectively improved PCN cell neurite growth and reduced TXNIP expression as well as interaction with NOD-like receptor pyrin domain-containing-3 (NLRP3) inflammasome, as determined by immunoblotting and immunoprecipitation. In our mouse model of extended hyperglycemic MCAO, i.a. verapamil (0.5 mg/kg) could attenuate neurological deficits induced by hyperglycemic stroke. This was associated with reduced microglial pro-inflammatory transition. This finding encourages pertinent studies in hyperglycemic patients undergoing thrombectomy where the robust reperfusion may exacerbate glucose toxicity.

9.
Transl Stroke Res ; 14(2): 211-237, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35596116

RESUMEN

Clinical evidence affirms physical exercise is effective in preventive and rehabilitation approaches for ischemic stroke. This sustainable efficacy is independent of cardiovascular risk factors and associates substantial reprogramming in circulating extracellular vesicles (EVs). The intricate journey of pluripotent exercise-induced EVs from parental cells to the whole-body and infiltration to cerebrovascular entity offers several mechanisms to reduce stroke incidence and injury or accelerate the subsequent recovery. This review delineates the potential roles of EVs as prospective effectors of exercise. The candidate miRNA and peptide cargo of exercise-induced EVs with both atheroprotective and neuroprotective characteristics are discussed, along with their presumed targets and pathway interactions. The existing literature provides solid ground to hypothesize that the rich vesicles link exercise to stroke prevention and rehabilitation. However, there are several open questions about the exercise stressors which may optimally regulate EVs kinetic and boost brain mitochondrial adaptations. This review represents a novel perspective on achieving brain fitness against stroke through transplantation of multi-potential EVs generated by multi-parental cells, which is exceptionally reachable in an exercising body.


Asunto(s)
Vesículas Extracelulares , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Accidente Cerebrovascular Isquémico/metabolismo , Estudios Prospectivos , Encéfalo/metabolismo , Accidente Cerebrovascular/terapia , Accidente Cerebrovascular/metabolismo , Vesículas Extracelulares/metabolismo
10.
Neuroscientist ; 29(6): 694-714, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35769016

RESUMEN

The neurovascular unit (NVU) refers to the functional building unit of the brain and the retina, where neurons, glia, and microvasculature orchestrate to meet the demand of the retina's and brain's function. Neurotrophins (NTs) are structural families of secreted proteins and are known for exerting neurotrophic effects on neuronal differentiation, survival, neurite outgrowth, synaptic formation, and plasticity. NTs include several molecules, such as nerve growth factor, brain-derived neurotrophic factor, NT-3, NT-4, and their precursors. Furthermore, NTs are involved in signaling pathways such as inflammation, apoptosis, and angiogenesis in a nonneuronal cell type. Interestingly, NTs and the precursors can bind and activate the p75 neurotrophin receptor (p75NTR) at low and high affinity. Mature NTs bind their cognate tropomyosin/tyrosine-regulated kinase receptors, crucial for maintenance and neuronal development in the brain and retina axis. Activation of p75NTR results in neuronal apoptosis and cell death, while tropomysin receptor kinase upregulation contributes to differentiation and cell growth. Recent findings indicate that modulation of NTs and their receptors contribute to neurovascular dysfunction in the NVU. Several chronic metabolic and acute ischemic diseases affect the NVU, including diabetic and ischemic retinopathy for the retina, as well as stroke, acute encephalitis, and traumatic brain injury for the brain. This work aims to review the current evidence through published literature studying the impact of NTs and their receptors, including the p75NTR receptor, on the injured and healthy brain-retina axis.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Receptor de Factor de Crecimiento Nervioso , Humanos , Receptor de Factor de Crecimiento Nervioso/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neuronas/metabolismo , Retina/metabolismo , Apoptosis/fisiología , Biología
11.
Exp Neurol ; 359: 114161, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35787888

RESUMEN

The precursor form of nerve growth factor (proNGF) is essential to maintain NGF survival signaling. ProNGF is also among endogenous ligands for p75 neurotrophin receptor (p75ntr). Mounting evidence implies that p75ntr signaling contributes to neural damage in ischemic stroke. The present study examines the therapeutic effect of the p75ntr modulator LM11A-31. Adult mice underwent transient distal middle cerebral artery occlusion (t-dMCAO) followed by LM11A-31 treatment (25 mg/kg, i.p., twice daily) either for 72 h post-injury (acute phase) or afterward till two weeks post-stroke (subacute phase). LM11A-31 reduced blood-brain barrier permeability, cerebral tissue injury, and sensorimotor function in the acute phase of stroke. Ischemic brain samples showed repressed proNGF/P75ntr signaling and Caspase 3 activation in LM11A-31 treated mice, where we observed less reactive microglia and IL-1ß production. LM11A-31 (20-80 nM) also mitigated neural injury induced by oxygen-glucose deprivation (OGD) in sandwich co-cultures of primary cortical neurons (PCN) and astrocytes. This concurred with JNK/PARP downregulation and reduced caspase-3 cleavage in the PCNs and was associated with repressed proNGF generation in astrocytes. Further in vitro experiments indicated human proNGF suppresses the pro-inflammatory phenotype in microglial cultures, as determined by a sharp decline in HMGB-1 production and moderate arginase-1 upregulation. Despite significant protection in acute stroke, LM11A-31 treatment did not improve cortical atrophy and sensorimotor function in the subacute phase. Our findings provide preclinical evidence supporting LM11A-31 as a promising therapy for acute stroke injury. Further investigations may elucidate if reduced astrocytic proNGF, an endogenous reservoir of pro-neurotrophins, may restrict the therapeutic window.


Asunto(s)
Receptor de Factor de Crecimiento Nervioso , Accidente Cerebrovascular , Ratones , Humanos , Animales , Receptor de Factor de Crecimiento Nervioso/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Astrocitos/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico
12.
J Pept Sci ; 18(2): 92-6, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22083687

RESUMEN

Dipeptidyl carboxypeptidase-I is an enzyme involved in the biological degradation of enkephalins. It has been suggested that C-terminal amidation of enkephalins enhances their resistance to dipeptidyl carboxypeptidase-I-mediated biodegradation. In this study, a novel [Met5]enkephalin amide (MEA) analogue [Met5]enkephalin (ME)-semicarbazide synthesized by another laboratory in our group was assessed for its antinociceptive effects compared with ME-ethylamide, MEA and ME, using tail flick test. To protect the administered drugs from biodegradation, rats were pretreated with peptidase inhibitors including amastatin, phosphoramidon and captopril. Then captopril (dipeptidyl carboxypeptidase-I inhibitor) was deleted from the peptidase inhibitors' combination for evaluating in vivo resistance of the synthetic drugs to dipeptidyl carboxypeptidase-I. According to the results, ME-semicarbazide and MEA were resistant enough to dipeptidyl carboxypeptidase-I to exert their strong antinociception following intrathecal administration even in the absence of captopril, whereas the antinociceptive effects produced by ME-ethylamide (10 nmol) were abolished in rats not pretreated with captopril, indicating that significant amounts of the ME-ethylamide were degraded by dipeptidyl carboxypeptidase-I. Replacement of the amide moiety of MEA with semicarbazide provides a new ME derivative, with high analgesic effects as well as more resistance to dipeptidyl carboxypeptidase-I-mediated biodegradation.


Asunto(s)
Analgésicos/farmacología , Carboxipeptidasas/metabolismo , Encefalina Metionina/análogos & derivados , Semicarbacidas/farmacología , Analgésicos/administración & dosificación , Analgésicos/metabolismo , Animales , Biotransformación , Captopril/farmacología , Carboxipeptidasas/antagonistas & inhibidores , Encefalina Metionina/administración & dosificación , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacocinética , Encefalina Metionina/farmacología , Glicopéptidos/farmacología , Hidrólisis , Inyecciones Espinales , Masculino , Nocicepción/efectos de los fármacos , Péptidos/farmacología , Inhibidores de Proteasas/farmacología , Ratas , Ratas Wistar , Semicarbacidas/farmacocinética
13.
Mol Neurobiol ; 59(10): 6281-6306, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35922728

RESUMEN

It is well acknowledged that neuroprotective effects of transplanted mesenchymal stem cells (MSCs) in ischemic stroke are attributed to their paracrine-mediated actions or bystander effects rather than to cell replacement in infarcted areas. This therapeutic plasticity is due to MSCs' ability to secrete a broad range of bioactive molecules including growth factors, trophic factors, cytokines, chemokines, and extracellular vesicles, overall known as the secretome. The secretome derivatives, such as conditioned medium (CM) or purified extracellular vesicles (EVs), exert remarkable advantages over MSC transplantation in stroke treating. Here, in this review, we used published information to provide an overview on the secretome composition of MSCs, underlying mechanisms of therapeutic effects of MSCs, and preclinical studies on MSC-derived products application in stroke. Furthermore, we discussed current advantages and challenges for successful bench-to-bedside translation.


Asunto(s)
Vesículas Extracelulares , Accidente Cerebrovascular Isquémico , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Accidente Cerebrovascular , Vesículas Extracelulares/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/terapia
14.
Mol Neurobiol ; 58(8): 3792-3804, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33847912

RESUMEN

Thrombolytic therapy has remained quite challenging in hyperglycemic patients for its association with poor prognosis and increased hemorrhagic conversions. We recently showed that tissue plasminogen activator (tPA)-induced cerebrovascular damage is associated with thioredoxin-interacting protein (TXNIP) upregulation, which has an established role in the detrimental effects of hyperglycemia. In the present work, we investigated whether verapamil, an established TXNIP inhibitor, may provide protection against hyperglycemic stroke and tPA-induced blood-brain barrier (BBB) disruption. Acute hyperglycemia was induced by intraperitoneal administration of 20% glucose, 15 min prior to transient middle cerebral artery occlusion (tMCAO). Verapamil (0.15 mg/kg) or saline was intravenously infused with tPA at hyperglycemic reperfusion, 1 h post tMCAO. After 24 h of ischemia/reperfusion (I/R), mice were assessed for neurobehavioral deficits followed by sacrifice and evaluation of brain infarct volume, edema, and microbleeding. Alterations in TXNIP, inflammatory mediators, and BBB markers were further analyzed using immunoblotting or immunostaining techniques. As adjunctive therapy, verapamil significantly reduced tPA-induced BBB leakage, matrix metalloproteinase 9 (MMP-9) upregulation, and tight junction protein deregulation, which resulted in lesser hemorrhagic conversions. Importantly, verapamil strongly reversed tPA-induced TXNIP/NLRP3 (NOD-like receptor pyrin domain-containing-3) inflammasome activation and reduced infarct volume. This concurred with a remarkable decrease in high-mobility group box protein 1 (HMGB-1) and nuclear factor kappa B (NF-κB) stimulation, leading to less priming of NLRP3 inflammasome. This preclinical study supports verapamil as a safe adjuvant that may complement thrombolytic therapy by inhibiting TXNIP's detrimental role in hyperglycemic stroke.


Asunto(s)
Proteínas Portadoras/metabolismo , Hiperglucemia/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Accidente Cerebrovascular/metabolismo , Tiorredoxinas/metabolismo , Activador de Tejido Plasminógeno/administración & dosificación , Verapamilo/administración & dosificación , Animales , Proteínas Portadoras/antagonistas & inhibidores , Quimioterapia Combinada , Fibrinolíticos/administración & dosificación , Hiperglucemia/tratamiento farmacológico , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Accidente Cerebrovascular/tratamiento farmacológico , Tiorredoxinas/antagonistas & inhibidores , Activador de Tejido Plasminógeno/toxicidad , Vasodilatadores/administración & dosificación
15.
Biomed Pharmacother ; 140: 111709, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34020250

RESUMEN

It is well accepted that the success of mesenchymal stem cells (MSCs) therapy against experimental stroke is mainly due to cellular paracrine manners rather than to replace lost tissue per se. Given such "bystander" effects, cell-free therapeutics manifest as a promising approach in regenerative medicine. Here we aimed at evaluating the effect of conditioned medium (CM) derived from human embryonic MSCs (hESC-MSC) on the neurological deficit, neurogenesis, and angiogenesis in experimental stroke. Adult male Wistar rats subjected to middle cerebral artery occlusion (MCAO), were treated with intracerebroventricular CM either one time (1 h post MCAO) or three times (1, 24, and 48 h post MCAO). Motor performance was assessed by the cylinder test on days 3 and 7. Cerebral samples were obtained for infarct size and molecular analysis on day 7 post-injury. Neurogenesis was evaluated by probing Nestin, Ki67, DCX, and Reelin transcripts and protein levels in the striatum, cortex, subventricular zone, and corpus callosum. The mRNA and protein expression of CD31 were also assessed in the striatum and cortical region to estimate angiogenesis post MCAO. Our findings demonstrate that CM treatment could significantly ameliorate neurological deficits and infarct volume in MCAO rats. Furthermore, ischemic stroke was associated with higher levels of neurogenesis and angiogenesis markers. Following treatment with CM, these markers were further potentiated in the brain regions. This study suggests that the therapeutic benefits of CM obtained from hESC-MSCs at least partly are mediated through improved neurogenesis and angiogenesis to accelerate the recovery of cerebral ischemia insult.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Células Madre Embrionarias Humanas/citología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Células Madre Mesenquimatosas , Neovascularización Fisiológica/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Proteína Doblecortina , Humanos , Infarto de la Arteria Cerebral Media/fisiopatología , Inyecciones Intraventriculares , Accidente Cerebrovascular Isquémico/fisiopatología , Masculino , Ratas Wistar , Proteína Reelina
16.
Mol Neurobiol ; 57(6): 2495-2508, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32172516

RESUMEN

Hyperglycemia has been shown to counterbalance the beneficial effects of tissue plasminogen activator (tPA) and increase the risk of intracerebral hemorrhage in ischemic stroke. Thioredoxin interacting protein (TXNIP) mediates hyperglycemia-induced oxidative damage and inflammation in the brain and reduces cerebral glucose uptake/utilization. We have recently reported that TXNIP-induced NLRP3 (NOD-like receptor pyrin domain-containing-3) inflammasome activation contributes to neuronal damage after ischemic stroke. Here, we tested the hypothesis that tPA induces TXNIP-NLRP3 inflammasome activation after ischemic stroke, in hyperglycemic mice. Acute hyperglycemia was induced in mice by intraperitoneal (IP) administration of a 20% glucose solution. This was followed by transient middle cerebral artery occlusion (t-MCAO), with or without intravenous (IV) tPA administered at reperfusion. The IV-tPA exacerbated hyperglycemia-induced neurological deficits, ipsilateral edema and hemorrhagic transformation, and accentuated peroxisome proliferator activated receptor-γ (PPAR-γ) upregulation and TXNIP/NLRP3 inflammasome activation after ischemic stroke. Higher expression of TXNIP in hyperglycemic t-MCAO animals augmented glucose transporter 1 (GLUT-1) downregulation and increased vascular endothelial growth factor-A (VEGF-A) expression/matrix metallopeptidase 9 (MMP-9) signaling, all of which result in blood brain barrier (BBB) disruption and increased permeability to endogenous immunoglobulin G (IgG). It was also associated with a discernible buildup of nitrotyrosine and accumulation of dysfunctional tight junction proteins: zonula occludens-1 (ZO-1), occludin and claudin-5. Moreover, tPA administration triggered activation of high mobility group box protein 1 (HMGB-1), nuclear factor kappa B (NF-κB), and tumor necrosis factor-α (TNF-α) expression in the ischemic penumbra of hyperglycemic animals. All of these observations suggest a powerful role for TXNIP-NLRP3 inflammasome activation in the tPA-induced toxicity seen with hyperglycemic stroke.


Asunto(s)
Proteínas Portadoras/metabolismo , Hiperglucemia/metabolismo , Inflamasomas/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Accidente Cerebrovascular/metabolismo , Tiorredoxinas/metabolismo , Activador de Tejido Plasminógeno/farmacología , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Transportador de Glucosa de Tipo 1/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/patología , Inflamasomas/metabolismo , Ratones , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/patología , Regulación hacia Arriba/efectos de los fármacos
17.
J Alzheimers Dis ; 68(1): 255-265, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30741672

RESUMEN

Alzheimer's disease (AD) is the most common form of age-associated dementia characterized by amyloid-ß plaques and neurofibrillary tangles. Recent studies have demonstrated that thioredoxin-interacting protein (TXNIP), an endogenous regulator of redox/glucose induced stress and inflammation, is now known to be upregulated in stroke, traumatic brain injury, diabetes and AD. We hypothesized that TXNIP overexpression sustains neurodegeneration through activation of the nucleotide binding and oligomerization domain-like receptor protein 3 in human AD brains. We analyzed TXNIP and the components of the NLRP3 inflammasome in the cortex of postmortem human brain samples by western blotting, real-time PCR, and immunohistochemical techniques in comparison with age-matched non-demented controls. Our results demonstrate that TXNIP protein as well as its mRNA levels in the cortex was significantly upregulated in AD compared to control brains. Moreover, using double immunofluorescence staining, TXNIP and interlukin-1ß (IL-1ß) were co-localized near Aß plaques and p-tau. These results suggest an association between TXNIP overexpression levels and AD pathogenesis. Further, a significant increased expression of cleaved caspase-1 and IL-1ß, the products of inflammasome activation, was detected in the cortex of AD brains. Together, these findings suggest that TXNIP, an upstream promising new therapeutic target, is a molecular link between inflammation and AD. The significant contribution of TXNIP to AD pathology suggests that strategies focusing on specific targeting of the TXNIP-NLRP3 inflammasome may lead to novel therapies for the management of AD and other age-related dementias.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Encéfalo/patología , Caspasa 1/metabolismo , Femenino , Humanos , Interleucina-1beta/metabolismo , Masculino , Persona de Mediana Edad , Placa Amiloide/metabolismo , Placa Amiloide/patología , Proteínas tau/metabolismo
18.
J Alzheimers Dis ; 66(3): 857-885, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30372683

RESUMEN

Empirical evidence indicates a strong association between insulin resistance and pathological alterations related to Alzheimer's disease (AD) in different cerebral regions. While cerebral insulin resistance is not essentially parallel with systemic metabolic derangements, type 2 diabetes mellitus (T2DM) has been established as a risk factor for AD. The circulating "toxic metabolites" emerging in metabolic syndrome may engage several biochemical pathways to promote oxidative stress and neuroinflammation leading to impair insulin function in the brain or "type 3 diabetes". Thioredoxin-interacting protein (TXNIP) as an intracellular amplifier of oxidative stress and inflammasome activation may presumably mediate central insulin resistance. Emerging data including those from our recent studies has demonstrated a sharp TXNIP upregulation in stroke, aging and AD and well underlining the significance of this hypothesis. With the main interest to illustrate TXNIP place in type 3 diabetes, the present review primarily briefs the potential mechanisms contributing to cerebral insulin resistance in a metabolically deranged environment. Then with a particular focus on plausible TXNIP functions to drive and associate with AD pathology, we present the most recent evidence supporting TXNIP as a promising therapeutic target in AD as an age-associated dementia.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Resistencia a la Insulina/fisiología , Síndrome Metabólico/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Estrés Oxidativo/fisiología
19.
Mol Neurobiol ; 55(10): 7900-7920, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29488135

RESUMEN

Neurological diseases, including acute attacks (e.g., ischemic stroke) and chronic neurodegenerative diseases (e.g., Alzheimer's disease), have always been one of the leading cause of morbidity and mortality worldwide. These debilitating diseases represent an enormous disease burden, not only in terms of health suffering but also in economic costs. Although the clinical presentations differ for these diseases, a growing body of evidence suggests that oxidative stress and inflammatory responses in brain tissue significantly contribute to their pathology. However, therapies attempting to prevent oxidative damage or inhibiting inflammation have shown little success. Identification and targeting endogenous "upstream" mediators that normalize such processes will lead to improve therapeutic strategy of these diseases. Thioredoxin-interacting protein (TXNIP) is an endogenous inhibitor of the thioredoxin (TRX) system, a major cellular thiol-reducing and antioxidant system. TXNIP regulating redox/glucose-induced stress and inflammation, now is known to get upregulated in stroke and other brain diseases, and represents a promising therapeutic target. In particular, there is growing evidence that glucose strongly induces TXNIP in multiple cell types, suggesting possible physiological roles of TXNIP in glucose metabolism. Recently, a significant body of literature has supported an essential role of TXNIP in the activation of the NOD-like receptor protein (NLRP3)-inflammasome, a well-established multi-molecular protein complex and a pivotal mediator of sterile inflammation. Accordingly, TXNIP has been postulated to reside centrally in detecting cellular damage and mediating inflammatory responses to tissue injury. The majority of recent studies have shown that pharmacological inhibition or genetic deletion of TXNIP is neuroprotective and able to reduce detrimental aspects of pathology following cerebrovascular and neurodegenerative diseases. Conspicuously, the mainstream of the emerging evidences is highlighting TXNIP link to damaging signals in endothelial cells. Thereby, here, we keep the trend to present the accumulative data on CNS diseases dealing with vascular integrity. This review aims to summarize evidence supporting the significant contribution of regulatory mechanisms of TXNIP with the development of brain diseases, explore pharmacological strategies of targeting TXNIP, and outline obstacles to be considered for efficient clinical translation.


Asunto(s)
Proteínas Portadoras/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Animales , Células Endoteliales/patología , Humanos , Inflamación/patología , Enfermedades Neurodegenerativas/patología , Estrés Oxidativo , Fibras de Estrés/patología
20.
J Neurotrauma ; 35(11): 1294-1303, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29295651

RESUMEN

Nucleotide oligomerization domain (NOD)-like receptor protein-3 (NLRP3) inflammasome may intimately contribute to sustaining damage after traumatic brain injury (TBI). This study aims to examine whether specific modulation of NLPR3 inflammasome by MCC950, a novel selective NLRP3 inhibitor, confers protection after experimental TBI. Unilateral cortical impact injury was induced in young adult C57BL/6 mice. MCC950 (50 mg/kg, intraperitoneally) or saline was administration at 1 and 3 h post-TBI. Animals were tested for neurological function and then sacrificed at 24 or 72 h post-TBI. Immunoblotting and histological analysis were performed to identify markers of NLRP3 inflammasome and proapoptotic activity in pericontusional areas of the brains at 24 or 72 h post-TBI. MCC950 treatment provided a significant improvement in neurological function and reduced cerebral edema in TBI animals. TBI upregulated NLRP3, apoptosis-associated speck-like adapter protein (ASC), cleaved caspase-1, and interlukein-1ß (IL-1ß) in the perilesional area. MCC950 efficiently repressed caspase-1 and IL-1ß with a transient effect on ASC and NLRP3 post-TBI. MCC950 treatment also provided protection against proapoptotic activation of poly (ADP-ribose) polymerase and caspase-3 associated with TBI. A concurrent inhibition of inflammasome priming was also detectable at the nuclear factor kappa B/p65 and caspase-1 level. Our findings support the implication of NLRP3 inflammasome in the pathogenesis of TBI and further suggests the therapeutic potential of MCC950.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Furanos/farmacología , Fármacos Neuroprotectores/farmacología , Receptores de Superficie Celular/antagonistas & inhibidores , Sulfonamidas/farmacología , Animales , Compuestos Heterocíclicos de 4 o más Anillos , Indenos , Ratones , Ratones Endogámicos C57BL , Sulfonas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA