Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
J Alzheimers Dis ; 98(4): 1349-1360, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38578894

RESUMEN

BACKGROUND: Background: Neurodegenerative diseases manifest behavioral dysfunction with disease progression. Intervention with neuropsychiatric drugs is part of most multi-drug treatment paradigms. However, only a fraction of patients responds to the treatments and those responding must deal with drug-drug interactions and tolerance issues generally attributed to off-target activities. Recent efforts have focused on the identification of underexplored targets and exploration of improved outcomes by treatment with selective molecular probes. Objective: As part of ongoing efforts to identify and validate additional targets amenable to therapeutic intervention, we examined levels of the serotonin 5-HT2b receptor (5-HT2bR) in Alzheimer's disease (AD) brains and the potential of a selective 5-HT2bR antagonist to counteract synaptic plasticity and memory damage induced by AD-related proteins, amyloid-ß, and tau. Methods: This work used a combination of biochemical, chemical biology, electrophysiological, and behavioral techniques. Biochemical methods included analysis of protein levels. Chemical biology methods included the use of an in vivo molecular probe MW071, a selective antagonist for the 5HT2bR. Electrophysiological methods included assessment of long-term potentiation (LTP), a type of synaptic plasticity thought to underlie memory formation. Behavioral studies investigated spatial memory and associative memory. Results: 5HT2bR levels are increased in brain specimens of AD patients compared to controls. 5HT2bR antagonist treatment rescued amyloid-ß and tau oligomer-induced impairment of synaptic plasticity and memory. Conclusions: The increased levels of 5HT-2bR in AD patient brains and the attenuation of disease-related synaptic and behavioral dysfunctions by MW071 treatment suggest that the 5HT-2bR is a molecular target worth pursuing as a potential therapeutic target.


Asunto(s)
Enfermedad de Alzheimer , Animales , Humanos , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Potenciación a Largo Plazo/fisiología , Trastornos de la Memoria/tratamiento farmacológico , Memoria Espacial
2.
Nat Commun ; 13(1): 1768, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35365657

RESUMEN

Myeloid cells play key roles in cancer immune suppression and tumor progression. In response to tumor derived factors, circulating monocytes and granulocytes extravasate into the tumor parenchyma where they stimulate angiogenesis, immune suppression and tumor progression. Chemokines, cytokines and interleukins stimulate PI3Kγ-mediated Rap1 activation, leading to conformational changes in integrin α4ß1 that promote myeloid cell extravasation and tumor inflammation Here we show that PI3Kγ activates a high molecular weight form of myosin light chain kinase, MLCK210, that promotes myosin-dependent Rap1 GTP loading, leading to integrin α4ß1 activation. Genetic or pharmacological inhibition of MLCK210 suppresses integrin α4ß1 activation, as well as tumor inflammation and progression. These results demonstrate a critical role for myeloid cell MLCK210 in tumor inflammation and serve as basis for the development of alternative approaches to develop immune oncology therapeutics.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Quinasa de Cadena Ligera de Miosina , Neoplasias , Adhesión Celular/fisiología , Humanos , Inflamación , Peso Molecular , Células Mieloides/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Neoplasias/genética
3.
PLoS One ; 17(1): e0262474, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35081152

RESUMEN

Alzheimer's disease (AD) is the leading cause of dementia in the elderly, but therapeutic options are lacking. Despite long being able to effectively treat the ill-effects of pathology present in various rodent models of AD, translation of these strategies to the clinic has so far been disappointing. One potential contributor to this situation is the fact that the vast majority of AD patients have other dementia-contributing comorbid pathologies, the most common of which are vascular in nature. This situation is modeled relatively infrequently in basic AD research, and almost never in preclinical studies. As part of our efforts to develop small molecule, anti-inflammatory therapeutics for neurological injury and disease, we have recently been exploring potentially promising treatments in preclinical multi-morbidity contexts. In the present study, we generated a mouse model of mixed amyloid and hyperhomocysteinemia (HHcy) pathology in which to test the efficacy of one of our anti-inflammatory compounds, MW151. HHcy can cause cerebrovascular damage and is an independent risk factor for both AD dementia and vascular contributions to cognitive impairment and dementia. We found that MW151 was able to partially rescue hippocampal-dependent spatial memory and learning deficits in this comorbidity context, and further, that the benefit is associated with a normalization of hippocampal metabolites detectable via magnetic resonance spectroscopy. These findings provide evidence that MW151 in particular, and potentially anti-inflammatory treatment more generally, may be beneficial in AD patients with comorbid vascular pathology.


Asunto(s)
Antiinflamatorios/uso terapéutico , Demencia/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Memoria/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Conducta Animal/efectos de los fármacos , Demencia/diagnóstico por imagen , Demencia/metabolismo , Modelos Animales de Enfermedad , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Imagen por Resonancia Magnética , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/diagnóstico por imagen , Trastornos de la Memoria/metabolismo , Ratones
4.
Biochim Biophys Acta Mol Cell Res ; 1868(11): 119104, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34302892

RESUMEN

BACKGROUND: Myosin light chain kinase (MLCK) is a Ca2+-calmodulin-dependent enzyme dedicated to phosphorylate and activate myosin II to provide force for various motile processes. In smooth muscle cells and many other cells, small MLCK (S-MLCK) is a major isoform. S-MLCK is an actomyosin-binding protein firmly attached to contractile machinery in smooth muscle cells. Still, it can leave this location and contribute to other cellular processes. However, molecular mechanisms for switching the S-MLCK subcellular localization have not been described. METHODS: Site-directed mutagenesis and in vitro protein phosphorylation were used to study functional roles of discrete in-vivo phosphorylated residues within the S-MLCK actin-binding domain. In vitro co-sedimentation analysis was applied to study the interaction of recombinant S-MLCK actin-binding fragment with filamentous actin. Subcellular distribution of phosphomimicking S-MLCK mutants was studied by fluorescent microscopy and differential cell extraction. RESULTS: Phosphorylation of S-MLCK actin-binding domain at Ser25 and/or Thr56 by proline-directed protein kinases or phosphomimicking these posttranslational modifications alters S-MLCK binding to actin filaments both in vitro and in cells, and induces S-MLCK subcellular translocation with no effect on the enzyme catalytic properties. CONCLUSIONS: Phosphorylation of the amino terminal actin-binding domain of S-MLCK renders differential subcellular targeting of the enzyme and may, thereby, contribute to a variety of context-dependent responses of S-MLCK to cellular and tissue stimuli. GENERAL SIGNIFICANCE: S-MLCK physiological function can potentially be modulated via phosphorylation of its actin recognition domain, a regulation distinct from the catalytic and calmodulin regulatory domains.


Asunto(s)
Quinasa de Cadena Ligera de Miosina/metabolismo , Animales , Células HEK293 , Humanos , Ratones , Células 3T3 NIH , Fosforilación , Proteínas Quinasas/metabolismo
5.
Clin Pharmacol Drug Dev ; 10(2): 131-143, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32255549

RESUMEN

MW01-6-189WH (MW189) is a novel central nervous system-penetrant small-molecule drug candidate that selectively attenuates stressor-induced proinflammatory cytokine overproduction and is efficacious in intracerebral hemorrhage and traumatic brain injury animal models. We report first-in-human, randomized, double-blind, placebo-controlled phase 1 studies to evaluate the safety, tolerability, and pharmacokinetics (PK) of single and multiple ascending intravenous doses of MW189 in healthy adult volunteers. MW189 was safe and well tolerated in single and multiple doses up to 0.25 mg/kg, with no clinically significant concerns. The most common drug-related treatment-emergent adverse event was infusion-site reactions, likely related to drug solution acidity. No clinically concerning changes were seen in vital signs, electrocardiograms, physical or neurological examinations, or safety laboratory results. PK analysis showed dose-proportional increases in plasma concentrations of MW189 after single or multiple doses, with approximately linear kinetics and no significant drug accumulation. Steady state was achieved by dose 3 for all dosing cohorts. A pilot pharmacodynamic study administering low-dose endotoxin to induce a systemic inflammatory response was done to evaluate the effects of a single intravenous dose of MW189 on plasma cytokine levels. MW189 treatment resulted in lower levels of the proinflammatory cytokine TNF-α and higher levels of the anti-inflammatory cytokine IL-10 compared with placebo treatment. The outcomes are consistent with the pharmacological mechanism of MW189. Overall, the safety profile, PK properties, and pharmacodynamic effect support further development of MW189 for patients with acute brain injury.


Asunto(s)
Antiinflamatorios/administración & dosificación , Inflamación/tratamiento farmacológico , Piperazinas/administración & dosificación , Piridazinas/administración & dosificación , Piridinas/administración & dosificación , Adulto , Antiinflamatorios/efectos adversos , Antiinflamatorios/farmacocinética , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Piperazinas/efectos adversos , Piperazinas/farmacocinética , Piridazinas/efectos adversos , Piridazinas/farmacocinética , Piridinas/efectos adversos , Piridinas/farmacocinética , Adulto Joven
7.
Cell Rep ; 29(12): 3885-3901.e5, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31851921

RESUMEN

Reduced expression of the survival motor neuron (SMN) protein causes the neurodegenerative disease spinal muscular atrophy (SMA). Here, we show that adeno-associated virus serotype 9 (AAV9)-mediated delivery of Stasimon-a gene encoding an endoplasmic reticulum (ER)-resident transmembrane protein regulated by SMN-improves motor function in a mouse model of SMA through multiple mechanisms. In proprioceptive neurons, Stasimon overexpression prevents the loss of afferent synapses on motor neurons and enhances sensory-motor neurotransmission. In motor neurons, Stasimon suppresses neurodegeneration by reducing phosphorylation of the tumor suppressor p53. Moreover, Stasimon deficiency converges on SMA-related mechanisms of p53 upregulation to induce phosphorylation of p53 through activation of p38 mitogen-activated protein kinase (MAPK), and pharmacological inhibition of this kinase prevents motor neuron death in SMA mice. These findings identify Stasimon dysfunction induced by SMN deficiency as an upstream driver of distinct cellular cascades that lead to synaptic loss and motor neuron degeneration, revealing a dual contribution of Stasimon to motor circuit pathology in SMA.


Asunto(s)
Proteínas de la Membrana/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/etiología , Células Receptoras Sensoriales/patología , Proteína 1 para la Supervivencia de la Neurona Motora/fisiología , Sinapsis/patología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Dependovirus/genética , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patología , Células Receptoras Sensoriales/metabolismo , Sinapsis/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
8.
Geroscience ; 41(5): 671-679, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31104189

RESUMEN

The blood-brain barrier (BBB) is critical in maintenance of brain homeostasis, and loss of its functional integrity is a key feature across a broad range of neurological insults. This includes both acute injuries such as traumatic brain injury and stroke, as well as more chronic pathologies associated with aging, such as vascular cognitive impairment and dementia (VCID). A specific form of myosin light chain kinase (MLCK210) is a major regulator of barrier integrity in general, including the BBB. Studies have demonstrated the potential of MLCK210 as a therapeutic target for peripheral disorders involving tissue barrier dysfunction, but less is known about its potential as a target for chronic neurologic disorders. We report here that genetic knockout (KO) of MLCK210 protects against cerebral microhemorrhages and neuroinflammation induced by chronic dietary hyperhomocysteinemia. Overall, the results are consistent with an accumulating body of evidence supporting MLCK210 as a potential therapeutic target for tissue barrier dysfunction and specifically implicate it in BBB dysfunction and neuroinflammation in a model of VCID.


Asunto(s)
Hemorragia Cerebral/prevención & control , Disfunción Cognitiva/metabolismo , Demencia/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Animales , Hemorragia Cerebral/patología , Modelos Animales de Enfermedad , Hiperhomocisteinemia , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lectinas/genética , Lectinas/metabolismo , Ratones Noqueados , Quinasa de Cadena Ligera de Miosina/genética , ARN Mensajero/metabolismo , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/metabolismo
9.
J Med Chem ; 62(11): 5298-5311, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-30978288

RESUMEN

The p38αMAPK is a serine/threonine protein kinase and a key node in the intracellular signaling networks that transduce and amplify stress signals into physiological changes. A preponderance of preclinical data and clinical observations established p38αMAPK as a brain drug discovery target involved in neuroinflammatory responses and synaptic dysfunction in multiple degenerative and neuropsychiatric brain disorders. We summarize the discovery of highly selective, brain-penetrant, small molecule p38αMAPK inhibitors that are efficacious in diverse animal models of neurologic disorders. A crystallography and pharmacoinformatic approach to fragment expansion enabled the discovery of an efficacious hit. The addition of secondary pharmacology screens to refinement delivered lead compounds with improved selectivity, appropriate pharmacodynamics, and efficacy. Safety considerations and additional secondary pharmacology screens drove optimization that delivered the drug candidate MW01-18-150SRM (MW150), currently in early stage clinical trials.


Asunto(s)
Encéfalo/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Encéfalo/efectos de los fármacos , Disfunción Cognitiva/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Enfermedades del Sistema Nervioso/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico
10.
Proc Natl Acad Sci U S A ; 115(43): E10245-E10254, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30297392

RESUMEN

Autism spectrum disorder (ASD) is a common neurobehavioral disorder with limited treatment options. Activation of p38 MAPK signaling networks has been identified in ASD, and p38 MAPK signaling elevates serotonin (5-HT) transporter (SERT) activity, effects mimicked by multiple, hyperfunctional SERT coding variants identified in ASD subjects. Mice expressing the most common of these variants (SERT Ala56) exhibit hyperserotonemia, a biomarker observed in ASD subjects, as well as p38 MAPK-dependent SERT hyperphosphorylation, elevated hippocampal 5-HT clearance, hypersensitivity of CNS 5-HT1A and 5-HT2A/2C receptors, and behavioral and gastrointestinal perturbations reminiscent of ASD. As the α-isoform of p38 MAPK drives SERT activation, we tested the hypothesis that CNS-penetrant, α-isoform-specific p38 MAPK inhibitors might normalize SERT Ala56 phenotypes. Strikingly, 1-week treatment of adult SERT Ala56 mice with MW150, a selective p38α MAPK inhibitor, normalized hippocampal 5-HT clearance, CNS 5-HT1A and 5-HT2A/2C receptor sensitivities, social interactions, and colonic motility. Conditional elimination of p38α MAPK in 5-HT neurons of SERT Ala56 mice restored 5-HT1A and 5-HT2A/2C receptor sensitivities as well as social interactions, mirroring effects of MW150. Our findings support ongoing p38α MAPK activity as an important determinant of the physiological and behavioral perturbations of SERT Ala56 mice and, more broadly, supports consideration of p38α MAPK inhibition as a potential treatment for core and comorbid phenotypes present in ASD subjects.


Asunto(s)
Encéfalo/metabolismo , Tracto Gastrointestinal/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Receptores de Serotonina 5-HT2/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Serotonina/metabolismo , Animales , Trastorno del Espectro Autista/metabolismo , Masculino , Ratones , Fenotipo , Transducción de Señal/fisiología
11.
Neurobiol Aging ; 70: 86-91, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30007168

RESUMEN

Neuroinflammation is a fundamental mechanism in Alzheimer's disease (AD) progression. The stress-induced activation of the p38α mitogen-activated protein kinase (MAPK) leads to increased production of proinflammatory cytokines and neurodegeneration. We investigated the effects of an isoform selective p38α MAPK inhibitor, MW01-18-150SRM (MW150), administered at 2.5 mg/kg/d (i.p.; 14 days) on early entorhinal cortex (EC) alterations in an AD mouse model carrying human mutations of the amyloid precursor protein (mhAPP). We used electrophysiological analyses with long-term potentiation induction in EC-containing brain slices and EC-relevant associative memory tasks. We found that MW150 was capable of rescuing long-term potentiation in 2-month old mhAPP mice. Acute delivery of MW150 to brain slices was similarly effective in rescuing long-term potentiation, with a comparable efficacy to that of the widely used multikinase inhibitor SB203580. MW150-treated mhAPP mice demonstrated improved ability to discriminate novel associations between objects and their position/context. Our findings suggest that the selective inhibition of the stress-activated p38α MAPK with MW150 can attenuate the EC dysfunctions associated with neuroinflammation in an early stage of AD progression.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Corteza Entorrinal/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridazinas/uso terapéutico , Piridinas/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Corteza Entorrinal/fisiopatología , Masculino , Memoria/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Piperazinas/farmacología , Isoformas de Proteínas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Piridazinas/farmacología , Piridinas/farmacología
12.
J Neuroinflammation ; 14(1): 75, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28381303

RESUMEN

BACKGROUND: Brain p38α mitogen-activated protein kinase (MAPK), a potential therapeutic target for cognitive dysfunction based on the neuroinflammation-synaptic dysfunction cycle of pathophysiology progression, offers an innovative pharmacological strategy via inhibiting the same activated target in both glia and neurons, thereby enhancing the possibility for efficacy. The highly selective, brain-penetrant p38αMAPK inhibitor MW150 attenuates cognitive dysfunction in two distinct Alzheimer's disease (AD)-relevant models and avoids the problems encountered with previous mixed-kinase inhibitor drug candidates. Therefore, it is essential that the glial effects of this CNS-active kinase inhibitor be addressed in order to anticipate future use in clinical investigations. METHODS: We explored the effects of MW150 on glial biology in the AD-relevant APP/PS1 knock-in (KI) mouse model where we previously showed efficacy in suppression of hippocampal-dependent associative and spatial memory deficits. MW150 (2.5 mg/kg/day) was administered daily to 11-12-month-old KI mice for 14 days, and levels of proinflammatory cytokines IL-1ß, TNFα, and IL-6 measured in homogenates of mouse cortex using ELISA. Glial markers IBA1, CD45, CD68, and GFAP were assessed by immunohistochemistry. Microglia and amyloid plaques were quantified by immunofluorescence staining followed by confocal imaging. Levels of soluble and insoluble of Aß40 and Aß42 were measured by ELISA. The studies of in vivo pharmacodynamic effects on markers of neuroinflammation were complemented by mechanistic studies in the murine microglia BV2 cell line, using live cell imaging techniques to monitor proliferation, migration, and phagocytosis activities. RESULTS: Intervention with MW150 in KI mice during the established therapeutic time window attenuated the increased levels of IL-1ß and TNFα but not IL-6. MW150 treatment also increased the IBA1+ microglia within a 15 µm radius of the amyloid plaques, without significantly affecting overall microglia or plaque volume. Levels of IBA1, CD45, CD68, GFAP, and Aß40 and Aß42 were not affected by MW150 treatment. MW150 did not significantly alter microglial migration, proliferation, or phagocytosis in BV2 cells. CONCLUSIONS: Our results demonstrate that MW150 at an efficacious dose can selectively modulate neuroinflammatory responses associated with pathology progression without pan-suppression of normal physiological functions of microglia.


Asunto(s)
Cognición/fisiología , Citocinas/biosíntesis , Microglía/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis , Animales , Línea Celular , Cognición/efectos de los fármacos , Citocinas/antagonistas & inhibidores , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/patología , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
13.
Alzheimers Res Ther ; 8(1): 54, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27974048

RESUMEN

BACKGROUND: Hyperphosphorylation and aggregation of tau protein are the pathological hallmarks of Alzheimer's disease and related tauopathies. We previously demonstrated that the microglial activation induces tau hyperphosphorylation and cognitive impairment via activation of p38 mitogen-activated protein kinase (p38 MAPK) in the hTau mouse model of tauopathy that was deficient for microglial fractalkine receptor CX3CR1. METHOD: We report an isoform-selective, brain-permeable, and orally bioavailable small molecule inhibitor of p38α MAPK (MW181) and its effects on tau phosphorylation in vitro and in hTau mice. RESULTS: First, pretreatment of mouse primary cortical neurons with MW181 completely blocked inflammation-induced p38α MAPK activation and AT8 (pS199/pS202) site tau phosphorylation, with the maximum effect peaking at 60-90 min after stimulation. Second, treatment of old (~20 months of age) hTau mice with MW181 (1 mg/kg body weight; 14 days via oral gavage) significantly reduced p38α MAPK activation compared with vehicle-administered hTau mice. This also resulted in a significant reduction in AT180 (pT231) site tau phosphorylation and Sarkosyl-insoluble tau aggregates. Third, MW181 treatment significantly increased synaptophysin protein expression and resulted in improved working memory. Fourth, MW181 administration reduced phosphorylated MAPK-activated protein kinase 2 (pMK2) and phosphorylated activating transcription factor 2 (pATF2), which are known substrates of p38α MAPK. Finally, MW181 reduced the expression of interferon-γ and interleukin-1ß. CONCLUSIONS: Taken together, these studies support p38α MAPK as a valid therapeutic target for the treatment of tauopathies.


Asunto(s)
Factor de Transcripción Activador 2/efectos de los fármacos , Interferón gamma/efectos de los fármacos , Interleucina-1beta/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Memoria a Corto Plazo/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Piridazinas/farmacología , Piridinas/farmacología , Tauopatías/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas tau/metabolismo , Animales , Conducta Animal , Corteza Cerebral/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Piridazinas/administración & dosificación , Piridinas/administración & dosificación , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas tau/efectos de los fármacos
14.
PLoS One ; 11(2): e0149451, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26871438

RESUMEN

A prevailing neuroinflammation hypothesis is that increased production of proinflammatory cytokines contributes to progressive neuropathology, secondary to the primary damage caused by a traumatic brain injury (TBI). In support of the hypothesis, post-injury interventions that inhibit the proinflammatory cytokine surge can attenuate the progressive pathology. However, other post-injury neuroinflammatory responses are key to endogenous recovery responses. Therefore, it is critical that pharmacological attenuation of detrimental or dysregulated neuroinflammatory processes avoid pan-suppression of inflammation. MW151 is a CNS-penetrant, small molecule experimental therapeutic that restores injury- or disease-induced overproduction of proinflammatory cytokines towards homeostasis without immunosuppression. Post-injury administration of MW151 in a closed head injury model of mild TBI suppressed acute cytokine up-regulation and downstream cognitive impairment. Here, we report results from a diffuse brain injury model in mice using midline fluid percussion. Low dose (0.5-5.0 mg/kg) administration of MW151 suppresses interleukin-1 beta (IL-1ß) levels in the cortex while sparing reactive microglia and astrocyte responses. To probe molecular mechanisms, we used live cell imaging of the BV-2 microglia cell line to demonstrate that MW151 does not affect proliferation, migration, or phagocytosis of the cells. Our results provide insight into the roles of glial responses to brain injury and indicate the feasibility of using appropriate dosing for selective therapeutic modulation of injurious IL-1ß increases while sparing other glial responses to injury.


Asunto(s)
Antiinflamatorios/uso terapéutico , Lesiones Encefálicas/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Interleucina-1beta/inmunología , Microglía/efectos de los fármacos , Pirimidinas/uso terapéutico , Animales , Antiinflamatorios/química , Encéfalo/inmunología , Encéfalo/patología , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/patología , Línea Celular , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Interleucina-1beta/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Microglía/inmunología , Microglía/patología , Fagocitosis/efectos de los fármacos , Pirimidinas/química
15.
Am J Physiol Gastrointest Liver Physiol ; 309(12): G988-97, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26514773

RESUMEN

Recent studies have implicated a pathogenic role for matrix metalloproteinases 9 (MMP-9) in inflammatory bowel disease. Although loss of epithelial barrier function has been shown to be a key pathogenic factor for the development of intestinal inflammation, the role of MMP-9 in intestinal barrier function remains unclear. The aim of this study was to investigate the role of MMP-9 in intestinal barrier function and intestinal inflammation. Wild-type (WT) and MMP-9(-/-) mice were subjected to experimental dextran sodium sulfate (DSS) colitis by administration of 3% DSS in drinking water for 7 days. The mouse colonic permeability was measured in vivo by recycling perfusion of the entire colon using fluorescently labeled dextran. The DSS-induced increase in the colonic permeability was accompanied by an increase in intestinal epithelial cell MMP-9 expression in WT mice. The DSS-induced increase in intestinal permeability and the severity of DSS colitis was found to be attenuated in MMP-9(-/-) mice. The colonic protein expression of myosin light chain kinase (MLCK) and phospho-MLC was found to be significantly increased after DSS administration in WT mice but not in MMP-9(-/-) mice. The DSS-induced increase in colonic permeability and colonic inflammation was attenuated in MLCK(-/-) mice and MLCK inhibitor ML-7-treated WT mice. The DSS-induced increase in colonic surface epithelial cell MLCK mRNA was abolished in MMP-9(-/-) mice. Lastly, increased MMP-9 protein expression was detected within the colonic surface epithelial cells in ulcerative colitis cases. These data suggest a role of MMP-9 in modulation of colonic epithelial permeability and inflammation via MLCK.


Asunto(s)
Colitis/enzimología , Colon/enzimología , Sulfato de Dextran , Mucosa Intestinal/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Uniones Estrechas/enzimología , Animales , Colitis/inducido químicamente , Colitis/genética , Colitis/patología , Colitis/prevención & control , Colon/efectos de los fármacos , Colon/patología , Modelos Animales de Enfermedad , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Metaloproteinasa 9 de la Matriz/análisis , Metaloproteinasa 9 de la Matriz/deficiencia , Metaloproteinasa 9 de la Matriz/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Permeabilidad , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Índice de Severidad de la Enfermedad , Transducción de Señal , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/patología , Factores de Tiempo
16.
J Neurosci ; 35(16): 6554-69, 2015 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-25904805

RESUMEN

Epidemiological studies have associated increased risk of Alzheimer's disease (AD)-related clinical symptoms with a medical history of head injury. Currently, little is known about pathophysiology mechanisms linked to this association. Persistent neuroinflammation is one outcome observed in patients after a single head injury. Neuroinflammation is also present early in relevant brain regions during AD pathology progression. In addition, previous mechanistic studies in animal models link neuroinflammation as a contributor to neuropathology and cognitive impairment in traumatic brain injury (TBI) or AD-related models. Therefore, we explored the potential interplay of neuroinflammatory responses in TBI and AD by analysis of the temporal neuroinflammatory changes after TBI in an AD model, the APP/PS1 knock-in (KI) mouse. Discrete temporal aspects of astrocyte, cytokine, and chemokine responses in the injured KI mice were delayed compared with the injured wild-type mice, with a peak neuroinflammatory response in the injured KI mice occurring at 7 d after injury. The neuroinflammatory responses were more persistent in the injured KI mice, leading to a chronic neuroinflammation. At late time points after injury, KI mice exhibited a significant impairment in radial arm water maze performance compared with sham KI mice or injured wild-type mice. Intervention with a small-molecule experimental therapeutic (MW151) that selectively attenuates proinflammatory cytokine production yielded improved cognitive behavior outcomes, consistent with a link between neuroinflammatory responses and altered risk for AD-associated pathology changes with head injury.


Asunto(s)
Envejecimiento , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Traumatismos Cerrados de la Cabeza/patología , Traumatismos Cerrados de la Cabeza/psicología , Mediadores de Inflamación/metabolismo , Enfermedad de Alzheimer/complicaciones , Precursor de Proteína beta-Amiloide/genética , Animales , Astrocitos/metabolismo , Lesiones Encefálicas , Quimiocinas/metabolismo , Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/psicología , Citocinas/metabolismo , Progresión de la Enfermedad , Femenino , Técnicas de Sustitución del Gen , Traumatismos Cerrados de la Cabeza/complicaciones , Traumatismos Cerrados de la Cabeza/fisiopatología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Microglía/metabolismo , Piridazinas/farmacología , Pirimidinas/farmacología
17.
J Neuroinflammation ; 12: 69, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25886256

RESUMEN

BACKGROUND: Evidence from clinical studies and preclinical animal models suggests that proinflammatory cytokine overproduction is a potential driving force for pathology progression in traumatic brain injury (TBI). This raises the possibility that selective targeting of the overactive cytokine response, a component of the neuroinflammation that contributes to neuronal dysfunction, may be a useful therapeutic approach. MW151 is a CNS-penetrant, small molecule experimental therapeutic that selectively restores injury- or disease-induced overproduction of proinflammatory cytokines towards homeostasis. We previously reported that MW151 administered post-injury (p.i.) is efficacious in a closed head injury (CHI) model of diffuse TBI in mice. Here we test dose dependence of MW151 to suppress the target mechanism (proinflammatory cytokine up-regulation), and explore the therapeutic window for MW151 efficacy. METHODS: We examined suppression of the acute cytokine surge when MW151 was administered at different times post-injury and the dose-dependence of cytokine suppression. We also tested a more prolonged treatment with MW151 over the first 7 days post-injury and measured the effects on cognitive impairment and glial activation. RESULTS: MW151 administered up to 6 h post-injury suppressed the acute cytokine surge, in a dose-dependent manner. Administration of MW151 over the first 7 days post-injury rescues the CHI-induced cognitive impairment and reduces glial activation in the focus area of the CHI. CONCLUSIONS: Our results identify a clinically relevant time window post-CHI during which MW151 effectively restores cytokine production back towards normal, with a resultant attenuation of downstream cognitive impairment.


Asunto(s)
Lesiones Encefálicas/complicaciones , Encéfalo/metabolismo , Trastornos del Conocimiento/etiología , Citocinas/metabolismo , Análisis de Varianza , Animales , Encéfalo/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Trastornos del Conocimiento/tratamiento farmacológico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Piridazinas/uso terapéutico , Pirimidinas/uso terapéutico , Factores de Tiempo
18.
ACS Chem Neurosci ; 6(4): 666-80, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25676389

RESUMEN

The first kinase inhibitor drug approval in 2001 initiated a remarkable decade of tyrosine kinase inhibitor drugs for oncology indications, but a void exists for serine/threonine protein kinase inhibitor drugs and central nervous system indications. Stress kinases are of special interest in neurological and neuropsychiatric disorders due to their involvement in synaptic dysfunction and complex disease susceptibility. Clinical and preclinical evidence implicates the stress related kinase p38αMAPK as a potential neurotherapeutic target, but isoform selective p38αMAPK inhibitor candidates are lacking and the mixed kinase inhibitor drugs that are promising in peripheral tissue disease indications have limitations for neurologic indications. Therefore, pursuit of the neurotherapeutic hypothesis requires kinase isoform selective inhibitors with appropriate neuropharmacology features. Synaptic dysfunction disorders offer a potential for enhanced pharmacological efficacy due to stress-induced activation of p38αMAPK in both neurons and glia, the interacting cellular components of the synaptic pathophysiological axis, to be modulated. We report a novel isoform selective p38αMAPK inhibitor, MW01-18-150SRM (=MW150), that is efficacious in suppression of hippocampal-dependent associative and spatial memory deficits in two distinct synaptic dysfunction mouse models. A synthetic scheme for biocompatible product and positive outcomes from pharmacological screens are presented. The high-resolution crystallographic structure of the p38αMAPK/MW150 complex documents active site binding, reveals a potential low energy conformation of the bound inhibitor, and suggests a structural explanation for MW150's exquisite target selectivity. As far as we are aware, MW150 is without precedent as an isoform selective p38MAPK inhibitor or as a kinase inhibitor capable of modulating in vivo stress related behavior.


Asunto(s)
Encéfalo/efectos de los fármacos , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Fármacos Neuroprotectores/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridazinas/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/psicología , Animales , Aprendizaje por Asociación/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Humanos , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/fisiopatología , Ratones Transgénicos , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/fisiología , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Estructura Molecular , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacocinética , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Ratas Sprague-Dawley , Memoria Espacial/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/fisiología
19.
J Neurosci ; 34(39): 13077-82, 2014 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-25253854

RESUMEN

Infantile neuronal ceroid lipofuscinosis (INCL) is an inherited neurodegenerative lysosomal storage disease (LSD) caused by a deficiency in palmitoyl protein thioesterase-1 (PPT1). Studies in Ppt1(-/-) mice demonstrate that glial activation is central to the pathogenesis of INCL. Astrocyte activation precedes neuronal loss, while cytokine upregulation associated with microglial reactivity occurs before and concurrent with neurodegeneration. Therefore, we hypothesized that cytokine cascades associated with neuroinflammation are important therapeutic targets for the treatment of INCL. MW01-2-151SRM (MW151) is a blood-brain barrier penetrant, small-molecule anti-neuroinflammatory that attenuates glial cytokine upregulation in models of neuroinflammation such as traumatic brain injury, Alzheimer's disease, and kainic acid toxicity. Thus, we used MW151, alone and in combination with CNS-directed, AAV-mediated gene therapy, as a possible treatment for INCL. MW151 alone decreased seizure susceptibility. When combined with AAV-mediated gene therapy, treated INCL mice had increased life spans, improved motor performance, and eradication of seizures. Combination-treated INCL mice also had decreased brain atrophy, astrocytosis, and microglial activation, as well as intermediary effects on cytokine upregulation. These data suggest that MW151 can attenuate seizure susceptibility but is most effective when used in conjunction with a therapy that targets the primary genetic defect.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Terapia Genética , Microglía/metabolismo , Lipofuscinosis Ceroideas Neuronales/terapia , Tioléster Hidrolasas/genética , Animales , Antiinflamatorios/farmacocinética , Antiinflamatorios/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Dependovirus/genética , Locomoción , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Piridazinas/farmacocinética , Piridazinas/uso terapéutico , Pirimidinas/farmacocinética , Pirimidinas/uso terapéutico , Convulsiones/terapia , Tioléster Hidrolasas/metabolismo
20.
PLoS One ; 9(8): e105191, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25127391

RESUMEN

The calcium/calmodulin-dependent protein kinase II (CaMKII) is abundant in the brain, where it makes important contributions to synaptic organization and homeostasis, including playing an essential role in synaptic plasticity and memory. Four genes encode isoforms of CaMKII (α, ß, δ, γ), with CaMKIIα and CaMKIIß highly expressed in the brain. Decades of molecular and cellular research, as well as the use of a large number of CaMKIIα mutant mouse lines, have provided insight into the pivotal roles of CaMKIIα in brain plasticity and cognition. However, less is known about the CaMKIIß isoform. We report the development and extensive behavioral and phenotypic characterization of a CaMKIIß knockout (KO) mouse. The CaMKIIß KO mouse was found to be smaller at weaning, with an altered body mass composition. The CaMKIIß KO mouse showed ataxia, impaired forelimb grip strength, and deficits in the rotorod, balance beam and running wheel tasks. Interestingly, the CaMKIIß KO mouse exhibited reduced anxiety in the elevated plus maze and open field tests. The CaMKIIß KO mouse also showed cognitive impairment in the novel object recognition task. Our results provide a comprehensive behavioral characterization of mice deficient in the ß isoform of CaMKII. The neurologic phenotypes and the construction of the genotype suggest the utility of this KO mouse strain for future studies of CaMKIIß in brain structure, function and development.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Animales , Ansiedad/enzimología , Ataxia/enzimología , Composición Corporal , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Trastornos del Conocimiento/enzimología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fuerza Muscular , Comportamiento de Nidificación , Patrones de Reconocimiento Fisiológico , Prueba de Desempeño de Rotación con Aceleración Constante , Aumento de Peso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...