Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(25): e2123265119, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35700359

RESUMEN

Metabolic aberrations impact the pathogenesis of multiple sclerosis (MS) and possibly can provide clues for new treatment strategies. Using untargeted metabolomics, we measured serum metabolites from 35 patients with relapsing-remitting multiple sclerosis (RRMS) and 14 healthy age-matched controls. Of 632 known metabolites detected, 60 were significantly altered in RRMS. Bioinformatics analysis identified an altered metabotype in patients with RRMS, represented by four changed metabolic pathways of glycerophospholipid, citrate cycle, sphingolipid, and pyruvate metabolism. Interestingly, the common upstream metabolic pathway feeding these four pathways is the glycolysis pathway. Real-time bioenergetic analysis of the patient-derived peripheral blood mononuclear cells showed enhanced glycolysis, supporting the altered metabolic state of immune cells. Experimental autoimmune encephalomyelitis mice treated with the glycolytic inhibitor 2-deoxy-D-glucose ameliorated the disease progression and inhibited the disease pathology significantly by promoting the antiinflammatory phenotype of monocytes/macrophage in the central nervous system. Our study provided a proof of principle for how a blood-based metabolomic approach using patient samples could lead to the identification of a therapeutic target for developing potential therapy.


Asunto(s)
Desarrollo de Medicamentos , Glucólisis , Metabolómica , Esclerosis Múltiple Recurrente-Remitente , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antimetabolitos/farmacología , Antimetabolitos/uso terapéutico , Desoxiglucosa/farmacología , Desoxiglucosa/uso terapéutico , Desarrollo de Medicamentos/métodos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Leucocitos Mononucleares/metabolismo , Ratones , Esclerosis Múltiple Recurrente-Remitente/sangre , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/metabolismo
2.
J Immunol ; 198(9): 3615-3626, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28341672

RESUMEN

Traumatic brain injury (TBI) is a major public health issue, producing significant patient mortality and poor long-term outcomes. Increasing evidence suggests an important, yet poorly defined, role for the immune system in the development of secondary neurologic injury over the days and weeks following a TBI. In this study, we tested the hypothesis that peripheral macrophage infiltration initiates long-lasting adaptive immune responses after TBI. Using a murine controlled cortical impact model, we used adoptive transfer, transgenic, and bone marrow chimera approaches to show increased infiltration and proinflammatory (classically activated [M1]) polarization of macrophages for up to 3 wk post-TBI. Monocytes purified from the injured brain stimulated the proliferation of naive T lymphocytes, enhanced the polarization of T effector cells (TH1/TH17), and decreased the production of regulatory T cells in an MLR. Similarly, elevated T effector cell polarization within blood and brain tissue was attenuated by myeloid cell depletion after TBI. Functionally, C3H/HeJ (TLR4 mutant) mice reversed M1 macrophage and TH1/TH17 polarization after TBI compared with C3H/OuJ (wild-type) mice. Moreover, brain monocytes isolated from C3H/HeJ mice were less potent stimulators of T lymphocyte proliferation and TH1/TH17 polarization compared with C3H/OuJ monocytes. Taken together, our data implicate TLR4-dependent, M1 macrophage trafficking/polarization into the CNS as a key mechanistic link between acute TBI and long-term, adaptive immune responses.


Asunto(s)
Lesiones Traumáticas del Encéfalo/inmunología , Macrófagos/fisiología , Células TH1/inmunología , Células Th17/inmunología , Receptor Toll-Like 4/genética , Inmunidad Adaptativa , Traslado Adoptivo , Animales , Diferenciación Celular/genética , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación/genética , Fenotipo
3.
Brain Behav Immun ; 68: 224-237, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29079445

RESUMEN

Inflammation is an important mediator of secondary neurological injury after traumatic brain injury (TBI). Endocannabinoids, endogenously produced arachidonate based lipids, have recently emerged as powerful anti-inflammatory compounds, yet the molecular and cellular mechanisms underlying these effects are poorly defined. Endocannabinoids are physiological ligands for two known cannabinoid receptors, CB1R and CB2R. In the present study, we hypothesized that selective activation of CB2R attenuates neuroinflammation and reduces neurovascular injury after TBI. Using a murine controlled cortical impact (CCI) model of TBI, we observed a dramatic upregulation of CB2R within infiltrating myeloid cells beginning at 72 h. Administration of the selective CB2R agonist, GP1a (1-5 mg/kg), attenuated pro-inflammatory M1 macrophage polarization, increased anti-inflammatory M2 polarization, reduced edema development, enhanced cerebral blood flow, and improved neurobehavioral outcomes after TBI. In contrast, the CB2R antagonist, AM630, worsened outcomes. Taken together, our findings support the development of selective CB2R agonists as a therapeutic strategy to improve TBI outcomes while avoiding the psychoactive effects of CB1R activation.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Indenos/farmacología , Pirazoles/farmacología , Receptor Cannabinoide CB2/metabolismo , Animales , Lesiones Encefálicas/complicaciones , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/metabolismo , Cannabinoides/uso terapéutico , Cannabis , Modelos Animales de Enfermedad , Endocannabinoides/uso terapéutico , Inflamación/complicaciones , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroinmunomodulación/fisiología , Receptor Cannabinoide CB2/fisiología , Receptores de Cannabinoides/metabolismo , Receptores de Cannabinoides/fisiología
4.
J Immunol ; 197(3): 747-60, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27354217

RESUMEN

The AMP-activated protein kinase, AMPK, is an energy-sensing, metabolic switch implicated in various metabolic disorders; however, its role in inflammation is not well defined. We have previously shown that loss of AMPK exacerbates experimental autoimmune encephalomyelitis (EAE) disease severity. In this study, we investigated the mechanism through which AMPK modulates inflammatory disease like EAE. AMPKα1 knockout (α1KO) mice with EAE showed severe demyelination and inflammation in the brain and spinal cord compared with wild-type due to higher expression of proinflammatory Th17 cytokines, including IL-17, IL-23, and IL-1ß, impaired blood-brain barrier integrity, and increased infiltration of inflammatory cells in the CNS. Infiltrated CD4 cells in the brains and spinal cords of α1KO with EAE were significantly higher compared with wild-type EAE and were characterized as IL-17 (IL-17 and GM-CSF double-positive) CD4 cells. Increased inflammatory response in α1KO mice was due to polarization of macrophages (Mϕ) to proinflammatory M1 type phenotype (IL-10(low)IL-23/IL-1ß/IL-6(high)), and these M1 Mϕ showed stronger capacity to induce allogenic as well as Ag-specific (myelin oligodendrocyte glycoprotein [MOG]35-55) T cell response. Mϕ from α1KO mice also enhanced the encephalitogenic property of MOG35-55-primed CD4 T cells in B6 mice. The increased encephalitogenic MOG-restricted CD4(+) T cells were due to an autocrine effect of IL-1ß/IL-23-mediated induction of IL-6 production in α1KO Mϕ, which in turn induce IL-17 and GM-CSF production in CD4 cells. Collectively, our data indicate that AMPK controls the inflammatory disease by regulating the M1 phenotype-Th17 axis in an animal model of multiple sclerosis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-17/inmunología , Macrófagos/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Citometría de Flujo , Immunoblotting , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Células Th17/inmunología
5.
Biochim Biophys Acta Mol Basis Dis ; 1863(10 Pt B): 2614-2626, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28533056

RESUMEN

Traumatic brain injury (TBI) is a leading cause of mortality and long-term morbidity worldwide. Despite decades of pre-clinical investigation, therapeutic strategies focused on acute neuroprotection failed to improve TBI outcomes. This lack of translational success has necessitated a reassessment of the optimal targets for intervention, including a heightened focus on secondary injury mechanisms. Chronic immune activation correlates with progressive neurodegeneration for decades after TBI; however, significant challenges remain in functionally and mechanistically defining immune activation after TBI. In this review, we explore the burgeoning evidence implicating the acute release of damage associated molecular patterns (DAMPs), such as adenosine 5'-triphosphate (ATP), high mobility group box protein 1 (HMGB1), S100 proteins, and hyaluronic acid in the initiation of progressive neurological injury, including white matter loss after TBI. The role that pattern recognition receptors, including toll-like receptor and purinergic receptors, play in progressive neurological injury after TBI is detailed. Finally, we provide support for the notion that resident and infiltrating macrophages are critical cellular targets linking acute DAMP release with adaptive immune responses and chronic injury after TBI. The therapeutic potential of targeting DAMPs and barriers to clinical translational, in the context of TBI patient management, are discussed.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Sustancia Blanca/metabolismo , Adenosina Trifosfato/inmunología , Adenosina Trifosfato/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/terapia , Proteína HMGB1/inmunología , Proteína HMGB1/metabolismo , Humanos , Ácido Hialurónico/inmunología , Ácido Hialurónico/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Receptores de Reconocimiento de Patrones/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Proteínas S100/inmunología , Proteínas S100/metabolismo , Sustancia Blanca/inmunología , Sustancia Blanca/patología
6.
J Biol Chem ; 290(52): 30697-712, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26546682

RESUMEN

We performed untargeted metabolomics in plasma of B6 mice with experimental autoimmune encephalitis (EAE) at the chronic phase of the disease in search of an altered metabolic pathway(s). Of 324 metabolites measured, 100 metabolites that mapped to various pathways (mainly lipids) linked to mitochondrial function, inflammation, and membrane stability were observed to be significantly altered between EAE and control (p < 0.05, false discovery rate <0.10). Bioinformatics analysis revealed six metabolic pathways being impacted and altered in EAE, including α-linolenic acid and linoleic acid metabolism (PUFA). The metabolites of PUFAs, including ω-3 and ω-6 fatty acids, are commonly decreased in mouse models of multiple sclerosis (MS) and in patients with MS. Daily oral administration of resolvin D1, a downstream metabolite of ω-3, decreased disease progression by suppressing autoreactive T cells and inducing an M2 phenotype of monocytes/macrophages and resident brain microglial cells. This study provides a proof of principle for the application of metabolomics to identify an endogenous metabolite(s) possessing drug-like properties, which is assessed for therapy in preclinical mouse models of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Esclerosis Múltiple/metabolismo , Plasma/metabolismo , Animales , Modelos Animales de Enfermedad , Ácidos Grasos Insaturados/química , Ácidos Grasos Insaturados/metabolismo , Femenino , Humanos , Redes y Vías Metabólicas , Metabolómica , Ratones , Plasma/química
7.
Arterioscler Thromb Vasc Biol ; 34(6): 1231-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24700124

RESUMEN

OBJECTIVE: Vascular cells, particularly endothelial cells, adopt aerobic glycolysis to generate energy to support cellular functions. The effect of endothelial glycolysis on angiogenesis remains unclear. 6-Phosphofructo-2-kinase/fructose-2, 6-bisphosphatase, isoform 3 (PFKFB3) is a critical enzyme for endothelial glycolysis. By blocking or deleting PFKFB3 in endothelial cells, we investigated the influence of endothelial glycolysis on angiogenesis both in vitro and in vivo. APPROACH AND RESULTS: Under hypoxic conditions or after treatment with angiogenic factors, endothelial PFKFB3 was upregulated both in vitro and in vivo. The knockdown or overexpression of PFKFB3 suppressed or accelerated endothelial proliferation and migration in vitro, respectively. Neonatal mice from a model of oxygen-induced retinopathy showed suppressed neovascular growth in the retina when endothelial PFKFB3 was genetically deleted or when the mice were treated with a PFKFB3 inhibitor. In addition, tumors implanted in mice deficient in endothelial PFKFB3 grew more slowly and were provided with less blood flow. A lower level of phosphorylated protein kinase B was observed in PFKFB3-knockdown endothelial cells, which was accompanied by a decrease in intracellular lactate. The addition of lactate to PFKFB3-knockdown cells rescued the suppression of endothelial proliferation and migration. CONCLUSIONS: The blockade or deletion of endothelial PFKFB3 decreases angiogenesis both in vitro and in vivo. Thus, PFKFB3 is a promising target for the reduction of endothelial glycolysis and its related pathological angiogenesis.


Asunto(s)
Células Endoteliales/fisiología , Neovascularización Patológica/etiología , Fosfofructoquinasa-2/fisiología , Animales , Proliferación Celular , Células Cultivadas , Femenino , Glucólisis , Humanos , Ácido Láctico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/fisiología , Factor A de Crecimiento Endotelial Vascular/farmacología
8.
Am J Physiol Regul Integr Comp Physiol ; 307(6): R693-703, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24990856

RESUMEN

20-Hydroxyeicosatetraenoic acid (20-HETE), Cyp4a-derived eicosanoid, is a lipid mediator that promotes tumor growth, as well as causing detrimental effects in cerebral circulation. We determined whether concurrent inhibition of cyclooxygenase-2 (COX-2) and 20-HETE affects colon tumor growth and ischemic stroke outcomes. The expression of Cyp4a and COXs and production of 20-HETE and PGE2 were determined in murine colon carcinoma (MC38) cells. We then examined the effects of combined treatment with rofecoxib, a potent COX-2 inhibitor, and HET0016, a potent Cyp4a inhibitor, on the growth and proliferation of MC38 cells. Subsequently, we tested the effects of HET0016 plus rofecoxib in MC38 tumor and ischemic stroke models. Cyp4a and COXs are highly expressed in MC38 cells. Respectively, HET0016 and rofecoxib inhibited 20-HETE and PGE2 formation in MC38 cells. Moreover, rofecoxib combined with HET0016 had greater inhibitory effects on the growth and proliferation of MC38 cells than did rofecoxib alone. Importantly, rofecoxib combined with HET0016 provided greater inhibition on tumor growth than did rofecoxib alone in MC38 tumor-bearing mice. Prolonged treatment with rofecoxib selectively induced circulating 20-HETE levels and caused cerebrovascular damage after ischemic stroke, whereas therapy with rofecoxib and HET0016 attenuated 20-HETE levels and reduced rofecoxib-induced cerebrovascular damage and stroke outcomes during anti-tumor therapy. Thus these results demonstrate that combination therapy with rofecoxib and HET0016 provides a new treatment of colon tumor, which can not only enhance the anti-tumor efficacy of rofecoxib, but also reduce rofecoxib-induced cerebrovascular damage and stroke outcomes.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Isquemia Encefálica/prevención & control , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Ciclooxigenasa 2/metabolismo , Ácidos Hidroxieicosatetraenoicos/antagonistas & inhibidores , Accidente Cerebrovascular/prevención & control , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Isquemia Encefálica/inducido químicamente , Isquemia Encefálica/enzimología , Línea Celular Tumoral , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Inhibidores de la Ciclooxigenasa 2/toxicidad , Citocromo P-450 CYP4A/antagonistas & inhibidores , Citocromo P-450 CYP4A/metabolismo , Dinoprostona/metabolismo , Humanos , Ácidos Hidroxieicosatetraenoicos/metabolismo , Lactonas/administración & dosificación , Lactonas/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Accidente Cerebrovascular/inducido químicamente , Accidente Cerebrovascular/enzimología , Sulfonas/administración & dosificación , Sulfonas/toxicidad , Factores de Tiempo , Carga Tumoral
9.
bioRxiv ; 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38979231

RESUMEN

Traumatic brain injury (TBI) is a significant public health concern characterized by a complex cascade of cellular events. TBI induces adenosine monophosphate-activated protein kinase (AMPK) dysfunction impairs energy balance activates inflammatory cytokines and leads to neuronal damage. AMPK is a key regulator of cellular energy homeostasis during inflammatory responses. Recent research has revealed its key role in modulating the inflammatory process in TBI. Following TBI the activation of AMPK can influence various important pathways and mechanisms including metabolic pathways and inflammatory signaling. Our study investigated the effects of post-TBI loss of AMPK function on functional outcomes inflammasome activation, and inflammatory cytokine production. Male C57BL/6 adult wild-type (WT) and AMPK knockout (AMPK-KO) mice were subjected to a controlled cortical impact (CCI) model of TBI or sham surgery. The mice were tested for behavioral impairment at 24 h post-TBI thereafter, mice were anesthetized, and their brains were quickly removed for histological and biochemical evaluation. In vitro we investigated inflammasome activation in mixed glial cells stimulated with lipopolysaccharides+ Interferon-gamma (LI) (0.1 µg/20 ng/ml LPS/IFNg) for 6 h to induce an inflammatory response. Estimating the nucleotide-binding domain, leucine-rich-containing family pyrin domain containing western blotting ELISA and qRT-PCR performed 3 (NLRP3) inflammasome activation and cytokine production. Our findings suggest that TBI leads to reduced AMPK phosphorylation in WT mice and that the loss of AMPK correlates with worsened behavioral deficits at 24 h post-TBI in AMPK-KO mice as compared to WT mice. Moreover compared with the WT mice AMPK-KO mice exhibit exacerbated NLRP3 inflammasome activation and increased expression of proinflammatory mediators such as IL-1b IL-6 TNF-a iNOS and Cox 2. These results align with the in vitro studies using brain glial cells under inflammatory conditions, demonstrating greater activation of inflammasome components in AMPK-KO mice than in WT mice. Our results highlighted the critical role of AMPK in TBI outcomes. We found that the absence of AMPK worsens behavioral deficits and heightens inflammasome-mediated inflammation thereby exacerbating brain injury after TBI. Restoring AMPK activity after TBI could be a promising therapeutic approach for alleviating TBI-related damage.

10.
Stroke ; 43(10): 2794-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22910893

RESUMEN

BACKGROUND AND PURPOSE: Remote ischemic conditioning is cardioprotective in myocardial infarction and neuroprotective in mechanical occlusion models of stroke. However, there is no report on its therapeutic potential in a physiologically relevant embolic stroke model (embolic middle cerebral artery occlusion) in combination with intravenous tissue-type plasminogen activator (tPA). METHODS: We tested remote ischemic perconditioning therapy (RIPerC) at 2 hours after embolic middle cerebral artery occlusion in the mouse with and without intravenous tPA at 4 hours. We assessed cerebral blood flow up to 6 hours, neurological deficits, injury size, and phosphorylation of Akt (Serine(473)) as a prosurvival signal in the ischemic hemisphere at 48 hours poststroke. RESULTS: RIPerC therapy alone improved the cerebral blood flow and neurological outcomes. tPA alone at 4 hours did not significantly improve the neurological outcome even after successful thrombolysis. Individual treatments with RIPerC and intravenous tPA reduced the infarct size (25.7% and 23.8%, respectively). Combination therapy of RIPerC and tPA resulted in additive effects in further improving the neurological outcome and reducing the infarct size (50%). All the therapeutic treatments upregulated phosphorylation of Akt in the ischemic hemisphere. CONCLUSIONS: RIPerC is effective alone after embolic middle cerebral artery occlusion and has additive effects in combination with intravenous tPA. RIPerC may be a simple, safe, and inexpensive combination therapy with intravenous tPA.


Asunto(s)
Infarto de la Arteria Cerebral Media/complicaciones , Precondicionamiento Isquémico/métodos , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/terapia , Terapia Trombolítica/métodos , Activador de Tejido Plasminógeno/uso terapéutico , Administración Intravenosa , Animales , Encéfalo/irrigación sanguínea , Encéfalo/fisiopatología , Terapia Combinada , Fibrinolíticos/administración & dosificación , Fibrinolíticos/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Flujo Sanguíneo Regional/fisiología , Accidente Cerebrovascular/patología , Factores de Tiempo , Activador de Tejido Plasminógeno/administración & dosificación , Resultado del Tratamiento
11.
Neurol Sci ; 33(5): 1011-20, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22170092

RESUMEN

Oxidative stress leads to complex biochemical alterations, and has been implicated in the progressive loss of learning and memory. Supplementing and boosting the endogenous antioxidant defense system could impede the progression of various types of neurodegeneration. In the present study, we have investigated the neuroprotective efficacy of a low-dose combination of certain promising and powerful natural antioxidants in an experimental model of cognitive impairment. Combined pretreatment with the extract of Nardosatchys jatamansi (N), crocetin (C) and selenium (Se) as sodium selenite (N, 200 mg/kg + C, 25 µg/kg + Se, 0.05 mg/kg body weight) for 15 days led to improved behavioral outcomes in streptozotocin (STZ)-induced cognitive impairment in rats. While intracerebroventricular (ICV) infusion of STZ resulted in the significant elevation of markers of oxidative stress and depletion of endogenous antioxidant defense system in the vehicle-pretreated group, these markers of oxidative stress and antioxidant enzymatic as well as non-enzymatic defense lines were attenuated in the group pretreated with the combination of antioxidants (NCSe). NCSe pretreatment markedly improved the performance of animals in passive avoidance test and Morris water maze (MWM) tasks, significantly reduced the level of TBARS, and elevated the content of glutathione and activities of antioxidant enzymes (glutathione peroxidase, glutathione-S-transferase and catalase). Our study reflects the synergistic potential of the above combination and concludes that a multimodal approach could be beneficial rather than a singular intervention.


Asunto(s)
Antioxidantes/administración & dosificación , Trastornos del Conocimiento , Nardostachys , Estrés Oxidativo/efectos de los fármacos , Fitoterapia/métodos , Animales , Carotenoides/administración & dosificación , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratas , Ratas Wistar , Selenio/administración & dosificación , Vitamina A/análogos & derivados
12.
Antioxidants (Basel) ; 11(10)2022 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-36290774

RESUMEN

The restoration of cerebral blood flow (CBF) to achieve brain tissue oxygenation (PbtO2) is the primary treatment for ischemic stroke, a significant cause of adult mortality and disability worldwide. Nitric oxide (NO) and its bioactive s-nitrosylated (SNO) reservoirs, such as s-nitrosoglutathione (GSNO), induce hypoxic vasodilation to enhance CBF during ischemia. The endogenous pool of SNOs/GSNO is enhanced via the activation of endothelial NO synthase (eNOS/NOS3) and by the suppression of class III alcohol dehydrogenase 5 (ADH5), also known as GSNO reductase (GSNOR). Remote ischemic conditioning (RIC), which augments NOS3 activity and SNO, is an emerging therapy in acute stroke. However, RIC has so far shown neutral effects in stroke clinical trials. As the majority of stroke patients are presented with endothelial dysfunctions and comorbidities, we tested the hypothesis that NOS3 dysfunction and diabetes will abolish the protective effects of RIC therapy in stroke, and the prior inhibition of GSNOR will turn RIC protective. Our data demonstrate that RIC during thrombotic stroke failed to enhance the CBF and the benefits of thrombolysis in NOS3 mutant (NOS3+/-) mice, a genetic model of NOS3 dysfunction. Interestingly, thrombotic stroke in diabetic mice enhanced the activity of GSNOR as early as 3 h post-stroke without decreasing the plasma nitrite (NO2-). In thrombotic stroke, neither a pharmacological inhibitor of GSNOR (GRI) nor RIC therapy alone was protective in diabetic mice. However, prior treatment with GRI followed by RIC enhanced the CBF and improved recovery. In a reperfused stroke model, the GRI-RIC combination therapy in diabetic mice augmented PbtO2, a translatory signature of successful microvascular reflow. In addition, RIC therapy unexpectedly increased the inflammatory markers at 6 h post-stroke in diabetic stroke that were downregulated in combination with GRI while improving the outcomes. Thus, we conclude that preexisting NOS3 dysfunctions due to comorbidities may neutralize the benefits of RIC in stroke, which can be turned protective in combination with GRI. Our findings may support the future clinical trial of RIC in comorbid stroke. Further studies are warranted to test and develop SNO reservoirs as the blood-associated biomarker to monitor the response and efficacy of RIC therapy in stroke.

13.
Neurochem Res ; 36(8): 1360-71, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21472457

RESUMEN

Experimental studies have demonstrated that oxidative stress and apoptosis play an important role in cerebral ischemic pathogenesis and may represent a target for treatment. The purpose of this study was to determine whether the quercetin dihydrate (Q) protects against cerebral ischemia neuronal damage. Male Wistar rats were subjected to transient middle cerebral artery occlusion (MCAO) for 2 h and reperfused for 72 h. Quercetin (30 mg/kg, i.p) was administrated 30 min before the onset of ischemia and after the ischemia at interval of 0, 24, 48, and 72 h. The administration of Q showed marked reduction in infarct size, reduced the neurological deficits in terms of behaviors, suppressed neuronal loss and diminished the p53 expression in MCAO rats. Q was found to be successful in upregulating the antioxidant status and lowering the TBARS level. Conversely, the elevated activity of poly (ADP-ribose) polymerase (PARP), and activity of caspase-3 in MCAO group was attenuated significantly in Q treated group when compared with MCAO group. Our study reveals that Q, as a powerful antioxidant, could prevent free radicals associated oxidative damage and morphological changes in the MCAO rats. Thus, it may have a therapeutic value for the treatment of stroke.


Asunto(s)
Ataque Isquémico Transitorio/tratamiento farmacológico , Ataque Isquémico Transitorio/patología , Estrés Oxidativo/efectos de los fármacos , Quercetina/farmacología , Quercetina/uso terapéutico , Daño por Reperfusión/prevención & control , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Glutatión/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Masculino , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ratas , Ratas Wistar , Prueba de Desempeño de Rotación con Aceleración Constante , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
14.
J Immunol ; 182(12): 8005-14, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19494326

RESUMEN

Experimental autoimmune encephalomyelitis (EAE) is a T cell-mediated autoimmune disease of the CNS. Metformin is the most widely used drug for diabetes and mediates its action via activating AMP-activated protein kinase (AMPK). We provide evidence that metformin attenuates the induction of EAE by restricting the infiltration of mononuclear cells into the CNS, down-regulating the expression of proinflammatory cytokines (IFN-gamma, TNF-alpha, IL-6, IL-17, and inducible NO synthase (iNOS)), cell adhesion molecules, matrix metalloproteinase 9, and chemokine (RANTES). Furthermore, the AMPK activity and lipids alterations (total phospholipids and in free fatty acids) were restored by metformin treatment in the CNS of treated EAE animals, suggesting the possible involvement of AMPK. Metformin activated AMPK in macrophages and thereby inhibited biosynthesis of phospholipids as well as neutral lipids and also down-regulated the expression of endotoxin (LPS)-induced proinflammatory cytokines and their mediators (iNOS and cyclooxygenase 2). It also attenuated IFN-gamma and IL-17-induced iNOS and cyclooxygenase 2 expression in RAW267.4 cells, further supporting its anti-inflammatory property. Metformin inhibited T cell-mediated immune responses including Ag-specific recall responses and production of Th1 or Th17 cytokines, while it induced the generation of IL-10 in spleen cells of treated EAE animals. Altogether these findings reveal that metformin may have a possible therapeutic value for the treatment of multiple sclerosis and other inflammatory diseases.


Asunto(s)
Sistema Nervioso Central/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Metformina/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Células Cultivadas , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Enfermedad Crónica , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Células TH1/efectos de los fármacos , Células TH1/inmunología
15.
Biomolecules ; 11(11)2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34827584

RESUMEN

The COVID-19 pandemic has escalated the occurrence of hypoxia including thrombotic stroke worldwide, for which nitric oxide (NO) therapy seems very promising and translatable. Therefore, various modes/routes of NO-delivery are now being tested in different clinical trials for safer, faster, and more effective interventions against ischemic insults. Intravenous (IV) infusion of S-Nitrosoglutathione (GSNO), the major endogenous molecular pool of NO, has been reported to protect against mechanical cerebral ischemia-reperfusion (IR); however, it has been never tested in any kind of "clinically" relevant thromboembolic stroke models with or without comorbidities and in combination with the thrombolytic reperfusion therapy. Moreover, "IV-effects" of higher dose of GSNO following IR-injury have been contradicted to augment stroke injury. Herein, we tested the hypothesis that nebulization of low-dose GSNO will not alter blood pressure (BP) and will mitigate stroke injury in diabetic mice via enhanced cerebral blood flow (CBF) and brain tissue oxygenation (PbtO2). GSNO-nebulization (200 µg/kgbwt) did not alter BP, but augmented the restoration of CBF, improved behavioral outcomes and reduced stroke injury. Moreover, GSNO-nebulization increased early reoxygenation of brain tissue/PbtO2 as measured at 6.5 h post-stroke following thrombolytic reperfusion, and enervated unwanted effects of late thrombolysis in diabetic stroke. We conclude that the GSNO-nebulization is safe and effective for enhancing collateral microvascular perfusion in the early hours following stroke. Hence, nebulized-GSNO therapy has the potential to be developed and translated into an affordable field therapy against ischemic events including strokes, particularly in developing countries with limited healthcare infrastructure.


Asunto(s)
Complicaciones de la Diabetes/tratamiento farmacológico , Diabetes Mellitus/tratamiento farmacológico , Hemorragia/prevención & control , S-Nitrosoglutatión/administración & dosificación , Accidente Cerebrovascular/complicaciones , Terapia Trombolítica/efectos adversos , Animales , Conducta Animal , Presión Sanguínea , Barrera Hematoencefálica , COVID-19/epidemiología , Hemorragia/complicaciones , Hipoxia , Infusiones Intravenosas , Flujometría por Láser-Doppler , Masculino , Ratones , Ratones Endogámicos C57BL , Microcirculación , Nebulizadores y Vaporizadores , Fármacos Neuroprotectores/farmacología , Perfusión , Daño por Reperfusión/tratamiento farmacológico , Riesgo , Estrés Mecánico
16.
Neuromolecular Med ; 23(3): 371-382, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33185833

RESUMEN

Traumatic optic neuropathy (TON) is characterized by visual dysfunction after indirect or direct injury to the optic nerve following blunt head trauma. TON is associated with increased oxidative stress and inflammation resulting in retinal ganglion cell (RGC) death. Remote ischemic post-conditioning (RIC) has been shown to enhance endogenous protective mechanisms in diverse disease models including stroke, vascular cognitive impairment (VCI), retinal injury and optic nerve injury. However, the protective mechanisms underlying the improvement of retinal function and RGC survival after RIC treatment remain unclear. Here, we hypothesized that RIC therapy may be protective following TON by preventing RGC death, oxidative insult and inflammation in the mouse retina. To carry out the study, mice were divided in three different groups (Control, TON and TON + RIC). We harvested retinal tissue 5 days after TON induction for western blotting and histochemical analysis. We observed increased TON-induced retinal cell death compared with controls by cleaved caspase-3 immunohistochemistry. Furthermore, the TON cohort demonstrated increased TUNEL positive cells which were significantly attenuated by RIC. Immunofluorescence data showed that oxidative stress markers dihydroethidium (DHE), NOX-2 and nitrotyrosine expression were elevated in the TON group relative to controls and RIC therapy significantly reduced the expression level of these markers. Next, we found that the proinflammatory cytokine TNF-α was increased and anti-inflammatory IL-10 was decreased in plasma of TON animals, and RIC therapy reversed this expression level. Interestingly, western blotting of retinal tissue showed that RGC marker Brn3a and tight junction proteins (ZO-1 and Occludin), and AMPKα1 expression were downregulated in the TON group compared to controls. However, RIC significantly increased the expression levels of these proteins. Together these data suggest that RIC therapy activates endogenous protective mechanisms which may attenuate TON-induced oxidative stress and inflammation, and improves BRB integrity.


Asunto(s)
Poscondicionamiento Isquémico , Traumatismos del Nervio Óptico/terapia , Adenilato Quinasa/biosíntesis , Adenilato Quinasa/genética , Animales , Barrera Hematorretinal , Caspasa 3/biosíntesis , Caspasa 3/genética , Muerte Celular , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/genética , Miembro Posterior/irrigación sanguínea , Interleucina-10/sangre , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , Modelos Animales , NADPH Oxidasa 2/análisis , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/prevención & control , Estrés Oxidativo , Células Ganglionares de la Retina/patología , Superóxidos/análisis , Factor de Transcripción Brn-3A/biosíntesis , Factor de Transcripción Brn-3A/genética , Factor de Necrosis Tumoral alfa/sangre , Tirosina/análogos & derivados , Tirosina/análisis
17.
Cond Med ; 4(3): 124-129, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34414362

RESUMEN

Remote ischemic conditioning (RIC) is a promising safe, feasible, and inexpensive treatment for acute stroke, both ischemic and hemorrhagic. It is applied with a blood pressure cuff on the limbs and is ideal for the prehospital setting. RIC is a form of preconditioning with similarities to physical exercise. Its mechanisms of action are multiple and include improvement of collateral cerebral blood flow (CBF) and RIC acts as a "collateral therapeutic". The increased CBF is likely related to nitric oxide synthase 3 in the endothelium and more importantly in circulating blood cells like the red blood cell. The RESIST clinical trial is a 1500 subject multicenter, randomized, sham-controlled trial of RIC in the prehospital setting in Denmark and should address the questions of whether RIC is safe and effective in acute stroke and whether the effect is mediated by an effect on nitric oxide/nitrite metabolism.

18.
JCI Insight ; 6(1)2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33427206

RESUMEN

The CNS is regarded as an immunoprivileged organ, evading routine immune surveillance; however, the coordinated development of immune responses profoundly influences outcomes after brain injury. Innate lymphoid cells (ILCs) are cytokine-producing cells that are critical for the initiation, modulation, and resolution of inflammation, but the functional relevance and mechanistic regulation of ILCs are unexplored after acute brain injury. We demonstrate increased proliferation of all ILC subtypes within the meninges for up to 1 year after experimental traumatic brain injury (TBI) while ILCs were present within resected dura and elevated within cerebrospinal fluid (CSF) of moderate-to-severe TBI patients. In line with energetic derangements after TBI, inhibition of the metabolic regulator, AMPK, increased meningeal ILC expansion, whereas AMPK activation suppressed proinflammatory ILC1/ILC3 and increased the frequency of IL-10-expressing ILC2 after TBI. Moreover, intracisternal administration of IL-33 activated AMPK, expanded ILC2, and suppressed ILC1 and ILC3 within the meninges of WT and Rag1-/- mice, but not Rag1-/- IL2rg-/- mice. Taken together, we identify AMPK as a brake on the expansion of proinflammatory, CNS-resident ILCs after brain injury. These findings establish a mechanistic framework whereby immunometabolic modulation of ILCs may direct the specificity, timing, and magnitude of cerebral immunity.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Lesiones Traumáticas del Encéfalo/enzimología , Lesiones Traumáticas del Encéfalo/inmunología , Inmunidad Innata , Linfocitos/inmunología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/deficiencia , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/inmunología , Adolescente , Adulto , Anciano , Animales , Lesiones Traumáticas del Encéfalo/líquido cefalorraquídeo , Modelos Animales de Enfermedad , Femenino , Humanos , Linfocitos/clasificación , Linfocitos/patología , Masculino , Meninges/inmunología , Meninges/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Adulto Joven
19.
Behav Pharmacol ; 21(5-6): 563-71, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20657266

RESUMEN

Increased oxidative stress is implicated in the pathogenesis of Parkinson's disease in which dopaminergic neurons are intrinsically susceptible to oxidative damage. Swiss albino mice were pretreated with Pycnogenol (PYC), an extract of Pinus maritime bark [20 mg/kg body weight, intraperitoneally (i.p.)] once daily for 15 days. Thereafter, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) (20 mg/kg body weight, intraperitoneally) was given four times at 2-hour intervals on 1 day only. Behaviours were altered in the MPTP group as compared with the vehicle-treated group and were restored in the PYC-pretreated MPTP group. The activity of antioxidant enzymes and the content of glutathione were significantly depleted in the MPTP-induced Parkinsonian group. The MPTP group pretreated with PYC showed significant protection of the activity of antioxidant enzymes and glutathione content when compared with the vehicle-treated MPTP group. A significantly elevated level of thiobarbituric acid reactive substances in the MPTP group was decreased significantly in the animals pretreated with PYC. An increase in the number of dopaminergic D2 receptors and decrease in the level of dopamine and its metabolite 3,4-dihydroxyphenyl acetic acid in the striatum were observed after MPTP injection, and restored significantly after PYC pretreatment. Thus, PYC may be used to prevent or reduce the deterioration caused by free radicals, thereby preventing subsequent behavioural and biochemical changes that occur in Parkinsonian mice.


Asunto(s)
Flavonoides/farmacología , Estrés Oxidativo/efectos de los fármacos , Trastornos Parkinsonianos/tratamiento farmacológico , Animales , Antioxidantes/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Flavonoides/aislamiento & purificación , Glutatión/metabolismo , Intoxicación por MPTP/tratamiento farmacológico , Intoxicación por MPTP/fisiopatología , Ratones , Trastornos Parkinsonianos/fisiopatología , Pinus/química , Extractos Vegetales/farmacología , Receptores de Dopamina D2/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
20.
Br J Pharmacol ; 177(11): 2466-2477, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31975437

RESUMEN

BACKGROUND AND PURPOSE: Intravenous thrombolysis (IVT) after stroke enhances C3a generation, which may abrogate the benefits of reperfusion. The C3aR antagonist SB290157 is neuroprotective following transient but not permanent middle cerebral artery occlusion (MCAo). SB290157 remains untested in thromboembolic (TE) models, which better approximate human stroke and also facilitate testing in combination with IVT. We hypothesized SB290157 would confer neuroprotection in TE stroke with and without "late" IVT. EXPERIMENTAL APPROACH: We used two different models of TE stroke to examine the efficacy of SB290157 alone and in combination with late IVT. We evaluated the benefit of SB290157 in attenuating post-ischaemic behavioural deficits, infarction, brain oedema and haemorrhage. KEY RESULTS: Plasma C3a was elevated 6 hr after TE stroke alongside increased cerebrovascular C3aR expression, which was sustained to 4 weeks. Increased C3aR expression also was visualized in human ischaemic brain. In a photothrombotic (PT) stroke model, which exhibits rapid spontaneous reperfusion, SB290157 given at 1 hr post-PT significantly improved neurofunction and reduced infarction at 48 hr. In an embolic (eMCAo) model, SB290157 administered at 2 hr improved histological and functional outcomes. Conversely, late IVT administered 4.5 hr post-eMCAo was ineffective likely due to increased haemorrhage and brain oedema. However, SB290157 administered prior to late IVT ameliorated haemorrhage and oedema and improved outcomes. CONCLUSIONS AND IMPLICATIONS: We conclude that SB290157 is safe and effective with and without late IVT following TE stroke. Therefore, C3a receptor antagonist therapy represents a promising candidate for clinical translation in stroke, particularly as an adjuvant to IVT.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular , Animales , Isquemia Encefálica/tratamiento farmacológico , Fibrinolíticos/uso terapéutico , Humanos , Ratones , Accidente Cerebrovascular/tratamiento farmacológico , Terapia Trombolítica , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA