Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Geroscience ; 44(5): 2491-2508, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35798912

RESUMEN

Insulin appears to exert salutary effects in the central nervous system (CNS). Thus, brain insulin resistance has been proposed to play a role in brain aging and dementia but is conceptually complex and unlikely to fit classic definitions established in peripheral tissues. Thus, we sought to characterize brain insulin responsiveness in young (4-5 months) and old (24 months) FBN male rats using a diverse set of assays to determine the extent to which insulin effects in the CNS are impaired with age. When performing hyperinsulinemic-euglycemic clamps in rats, intracerebroventricular (ICV) infusion of insulin in old animals improved peripheral insulin sensitivity by nearly two-fold over old controls and comparable to young rats, suggesting preservation of this insulin-triggered response in aging per se (p < 0.05). We next used an imaging-based approach by comparing ICV vehicle versus insulin and performed resting state functional magnetic resonance imaging (rs-fMRI) to evaluate age- and insulin-related changes in network connectivity within the default mode network. In aging, lower connectivity between the mesial temporal (MT) region and other areas, as well as reduced MT signal complexity, was observed in old rats, which correlated with greater cognitive deficits in old. Despite these stark differences, ICV insulin failed to elicit any significant alteration to the BOLD signal in young rats, while a significant deviation of the BOLD signal was observed in older animals, characterized by augmentation in regions of the septal nucleus and hypothalamus, and reduction in thalamus and nucleus accumbens. In contrast, ex vivo stimulation of hippocampus with 10 nM insulin revealed increased Akt activation in young (p < 0.05), but not old rats. Despite similar circulating levels of insulin and IGF-1, cerebrospinal fluid concentrations of these ligands were reduced with age. Thus, these data highlight the complexity of capturing brain insulin action and demonstrate preserved or heightened brain responses to insulin with age, despite dampened canonical signaling, thereby suggesting impaired CNS input of these ligands may be a feature of reduced brain insulin action, providing further rationale for CNS replacement strategies.


Asunto(s)
Resistencia a la Insulina , Insulina , Masculino , Ratas , Animales , Encéfalo , Envejecimiento/fisiología , Resistencia a la Insulina/fisiología , Hipocampo/fisiología
2.
Proc Natl Acad Sci U S A ; 118(40)2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34583988

RESUMEN

RNA polymerase (Pol) III synthesizes abundant short noncoding RNAs that have essential functions in protein synthesis, secretion, and other processes. Despite the ubiquitous functions of these RNAs, mutations in Pol III subunits cause Pol III-related leukodystrophy, an early-onset neurodegenerative disease. The basis of this neural sensitivity and the mechanisms of disease pathogenesis are unknown. Here we show that mice expressing pathogenic mutations in the largest Pol III subunit, Polr3a, specifically in Olig2-expressing cells, have impaired growth and developmental delay, deficits in cognitive, sensory, and fine sensorimotor function, and hypomyelination in multiple regions of the cerebrum and spinal cord. These phenotypes reflect a subset of clinical features seen in patients. In contrast, the gross motor defects and cerebellar hypomyelination that are common features of severely affected patients are absent in the mice, suggesting a relatively mild form of the disease in this conditional model. Our results show that disease pathogenesis in the mice involves defects that reduce both the number of mature myelinating oligodendrocytes and the ability of these cells to produce a myelin sheath of normal thickness. The findings suggest unique sensitivities of oligodendrogenesis and myelination to perturbations of Pol III transcription.


Asunto(s)
Enfermedades Desmielinizantes/fisiopatología , Mutación , ARN Polimerasa III/genética , Animales , Enfermedades Desmielinizantes/genética , Crecimiento , Humanos , Masculino , Ratones , Ratones Mutantes
3.
J Exp Med ; 218(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33045060

RESUMEN

Sickle cell disease (SCD) is a common hereditary hematologic disorder. SCD patients suffer from acute vaso-occlusive episodes (VOEs), chronic organ damage, and premature death, with few therapeutic options. Although severe pain is a major clinical manifestation of SCD, it remains unknown whether nociception plays a role in SCD pathogenesis. To address this question, we generated nociceptor-deficient SCD mice and found, unexpectedly, that the absence of nociception led to more severe and more lethal VOE, indicating that somatosensory nerves protect SCD mice from VOE. Mechanistically, the beneficial effects of sensory nerves were induced by the neuropeptide calcitonin gene-related peptide (CGRP), which acted on hematopoietic cells. Additionally, oral capsaicin consumption, which can activate somatosensory nerves by binding to TRPV1, dramatically alleviated acute VOE and significantly prevented chronic liver and kidney damage in SCD mice. Thus, the manipulation of nociception may provide a promising approach to treat SCD.


Asunto(s)
Anemia de Células Falciformes , Capsaicina/farmacología , Nociceptores/metabolismo , Células Receptoras Sensoriales/metabolismo , Enfermedades Vasculares , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/metabolismo , Animales , Péptido Relacionado con Gen de Calcitonina/genética , Péptido Relacionado con Gen de Calcitonina/metabolismo , Humanos , Hepatopatías/genética , Hepatopatías/metabolismo , Hepatopatías/prevención & control , Ratones , Ratones Noqueados , Dolor/tratamiento farmacológico , Dolor/genética , Dolor/metabolismo , Dolor/patología , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/prevención & control , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Enfermedades Vasculares/tratamiento farmacológico , Enfermedades Vasculares/genética , Enfermedades Vasculares/metabolismo
4.
Glia ; 69(3): 779-791, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33079443

RESUMEN

Adult onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) is a dementia resulting from dominantly inherited CSF1R inactivating mutations. The Csf1r+/- mouse mimics ALSP symptoms and pathology. Csf1r is mainly expressed in microglia, but also in cortical layer V neurons that are gradually lost in Csf1r+/- mice with age. We therefore examined whether microglial or neuronal Csf1r loss caused neurodegeneration in Csf1r+/- mice. The behavioral deficits, pathologies and elevation of Csf2 expression contributing to disease, previously described in the Csf1r+/- ALSP mouse, were reproduced by microglial deletion (MCsf1rhet mice), but not by neural deletion. Furthermore, increased Csf2 expression by callosal astrocytes, oligodendrocytes, and microglia was observed in Csf1r+/- mice and, in MCsf1rhet mice, the densities of these three cell types were increased in supraventricular patches displaying activated microglia, an early site of disease pathology. These data confirm that ALSP is a primary microgliopathy and inform future therapeutic and experimental approaches.


Asunto(s)
Enfermedades Desmielinizantes , Leucoencefalopatías , Enfermedades Neurodegenerativas , Animales , Leucoencefalopatías/genética , Ratones , Microglía , Neuroglía , Proteínas Tirosina Quinasas Receptoras , Receptores del Factor Estimulante de Colonias , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética
5.
Cell Rep ; 30(9): 3004-3019.e5, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32130903

RESUMEN

CSF-1R haploinsufficiency causes adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP). Previous studies in the Csf1r+/- mouse model of ALSP hypothesized a central role of elevated cerebral Csf2 expression. Here, we show that monoallelic deletion of Csf2 rescues most behavioral deficits and histopathological changes in Csf1r+/- mice by preventing microgliosis and eliminating most microglial transcriptomic alterations, including those indicative of oxidative stress and demyelination. We also show elevation of Csf2 transcripts and of several CSF-2 downstream targets in the brains of ALSP patients, demonstrating that the mechanisms identified in the mouse model are functional in humans. Our data provide insights into the mechanisms underlying ALSP. Because increased CSF2 levels and decreased microglial Csf1r expression have also been reported in Alzheimer's disease and multiple sclerosis, we suggest that the unbalanced CSF-1R/CSF-2 signaling we describe in the present study may contribute to the pathogenesis of other neurodegenerative conditions.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Microglía/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Transducción de Señal , Alelos , Animales , Antiinflamatorios/metabolismo , Antioxidantes/metabolismo , Atrofia , Depresión/prevención & control , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Gliosis/patología , Heterocigoto , Homeostasis , Humanos , Leucocitos/patología , Leucoencefalopatías/genética , Leucoencefalopatías/patología , Leucoencefalopatías/fisiopatología , Ratones Endogámicos C57BL , Microglía/patología , Actividad Motora , Vaina de Mielina/patología , Bulbo Olfatorio/patología , Bulbo Olfatorio/fisiopatología , Estrés Oxidativo , Fenotipo , Receptor de Factor Estimulante de Colonias de Macrófagos/deficiencia , Memoria Espacial , Transcriptoma/genética , Sustancia Blanca/patología , Sustancia Blanca/fisiopatología
6.
Geroscience ; 41(2): 185-208, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31076997

RESUMEN

Disruptions in growth hormone/insulin-like growth factor-1 (GH/IGF-1) signaling have been linked to improved longevity in mice and humans. Nevertheless, while IGF-1 levels are associated with increased cancer risk, they have been paradoxically implicated with protection from other age-related conditions, particularly in the brain, suggesting that strategies aimed at selectively increasing central IGF-1 action may have favorable effects on aging. To test this hypothesis, we generated inducible, brain-specific (TRE-IGF-1 × Camk2a-tTA) IGF-1 (bIGF-1) overexpression mice and studied effects on healthspan. Doxycycline was removed from the diet at 12 weeks old to permit post-development brain IGF-1 overexpression, and animals were monitored up to 24 months. Brain IGF-1 levels were increased approximately twofold in bIGF-1 mice, along with greater brain weights, volume, and myelin density (P < 0.05). Age-related changes in rotarod performance, exercise capacity, depressive-like behavior, and hippocampal gliosis were all attenuated specifically in bIGF-1 male mice (P < 0.05). However, chronic brain IGF-1 failed to prevent declines in cognitive function or neurovascular coupling. Therefore, we performed a short-term intranasal (IN) treatment of either IGF-1 or saline in 24-month-old male C57BL/6 mice and found that IN IGF-1 treatment tended to reduce depressive (P = 0.09) and anxiety-like behavior (P = 0.08) and improve motor coordination (P = 0.07) and unlike transgenic mice improved motor learning (P < 0.05) and visuospatial and working memory (P < 0.05). These data highlight important sex differences in how brain IGF-1 action impacts healthspan and suggest that translational approaches that target IGF-1 centrally can restore cognitive function, a possibility that should be explored as a strategy to combat age-related cognitive decline.


Asunto(s)
Envejecimiento/genética , Disfunción Cognitiva/genética , Regulación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/genética , Trastornos Psicomotores/genética , Animales , Modelos Animales de Enfermedad , Femenino , Longevidad/genética , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Distribución Aleatoria , Corteza Sensoriomotora , Transducción de Señal
7.
J Neurosci ; 39(10): 1892-1909, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30626701

RESUMEN

Emerging studies are providing compelling evidence that the pathogenesis of Huntington's disease (HD), a neurodegenerative disorder with frequent midlife onset, encompasses developmental components. Moreover, our previous studies using a hypomorphic model targeting huntingtin during the neurodevelopmental period indicated that loss-of-function mechanisms account for this pathogenic developmental component (Arteaga-Bracho et al., 2016). In the present study, we specifically ascertained the roles of subpallial lineage species in eliciting the previously observed HD-like phenotypes. Accordingly, we used the Cre-loxP system to conditionally ablate the murine huntingtin gene (Httflx) in cells expressing the subpallial patterning markers Gsx2 (Gsx2-Cre) or Nkx2.1 (Nkx2.1-Cre) in Httflx mice of both sexes. These genetic manipulations elicited anxiety-like behaviors, hyperkinetic locomotion, age-dependent motor deficits, and weight loss in both Httflx;Gsx2-Cre and Httflx;Nkx2.1-Cre mice. In addition, these strains displayed unique but complementary spatial patterns of basal ganglia degeneration that are strikingly reminiscent of those seen in human cases of HD. Furthermore, we observed early deficits of somatostatin-positive and Reelin-positive interneurons in both Htt subpallial null strains, as well as early increases of cholinergic interneurons, Foxp2+ arkypallidal neurons, and incipient deficits with age-dependent loss of parvalbumin-positive neurons in Httflx;Nkx2.1-Cre mice. Overall, our findings indicate that selective loss-of-huntingtin function in subpallial lineages differentially disrupts the number, complement, and survival of forebrain interneurons and globus pallidus GABAergic neurons, thereby leading to the development of key neurological hallmarks of HD during adult life. Our findings have important implications for the establishment and deployment of neural circuitries and the integrity of network reserve in health and disease.SIGNIFICANCE STATEMENT Huntington's disease (HD) is a progressive degenerative disorder caused by aberrant trinucleotide expansion in the huntingtin gene. Mechanistically, this mutation involves both loss- and gain-of-function mechanisms affecting a broad array of cellular and molecular processes. Although huntingtin is widely expressed during adult life, the mutant protein only causes the demise of selective neuronal subtypes. The mechanisms accounting for this differential vulnerability remain elusive. In this study, we have demonstrated that loss-of-huntingtin function in subpallial lineages not only differentially disrupts distinct interneuron species early in life, but also leads to a pattern of neurological deficits that are reminiscent of HD. This work suggests that early disruption of selective neuronal subtypes may account for the profiles of enhanced regional cellular vulnerability to death in HD.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Proteína Huntingtina/fisiología , Enfermedad de Huntington/fisiopatología , Interneuronas/fisiología , Neuronas/fisiología , Animales , Ansiedad/fisiopatología , Conducta Animal , Encéfalo/patología , Cuerpo Estriado/crecimiento & desarrollo , Cuerpo Estriado/patología , Femenino , Globo Pálido/crecimiento & desarrollo , Globo Pálido/patología , Proteína Huntingtina/genética , Enfermedad de Huntington/patología , Enfermedad de Huntington/psicología , Interneuronas/ultraestructura , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Corteza Motora/crecimiento & desarrollo , Corteza Motora/patología , Neuronas/ultraestructura , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/patología , Proteína Reelina
8.
Neurobiol Learn Mem ; 165: 106780, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-29307548

RESUMEN

Behavioral neuroscience research incorporates the identical high level of meticulous methodologies and exacting attention to detail as all other scientific disciplines. To achieve maximal rigor and reproducibility of findings, well-trained investigators employ a variety of established best practices. Here we explicate some of the requirements for rigorous experimental design and accurate data analysis in conducting mouse and rat behavioral tests. Novel object recognition is used as an example of a cognitive assay which has been conducted successfully with a range of methods, all based on common principles of appropriate procedures, controls, and statistics. Directors of Rodent Core facilities within Intellectual and Developmental Disabilities Research Centers contribute key aspects of their own novel object recognition protocols, offering insights into essential similarities and less-critical differences. Literature cited in this review article will lead the interested reader to source papers that provide step-by-step protocols which illustrate optimized methods for many standard rodent behavioral assays. Adhering to best practices in behavioral neuroscience will enhance the value of animal models for the multiple goals of understanding biological mechanisms, evaluating consequences of genetic mutations, and discovering efficacious therapeutics.


Asunto(s)
Investigación Conductal/métodos , Ratones/psicología , Ratas/psicología , Animales , Investigación Conductal/normas , Reproducibilidad de los Resultados , Proyectos de Investigación
9.
Neurobiol Learn Mem ; 165: 106867, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-29772390

RESUMEN

Christianson syndrome (CS) is a recently described rare neurogenetic disorder presenting early in life with a broad range of neurological symptoms, including severe intellectual disability with nonverbal status, hyperactivity, epilepsy, and progressive ataxia due to cerebellar atrophy. CS is due to loss-of-function mutations in SLC9A6, encoding NHE6, a sodium-hydrogen exchanger involved in the regulation of early endosomal pH. Here we review what is currently known about the neuropathogenesis of CS, based on insights from experimental models, which to date have focused on mechanisms that affect the CNS, specifically the brain. In addition, parental reports of sensory disturbances in their children with CS, including an apparent insensitivity to pain, led us to explore sensory function and related neuropathology in Slc9a6 KO mice. We present new data showing sensory deficits in Slc9a6 KO mice, which had reduced behavioral responses to noxious thermal and mechanical stimuli (Hargreaves and Von Frey assays, respectively) compared to wild type (WT) littermates. Immunohistochemical and ultrastructural analysis of the spinal cord and peripheral nervous system revealed intracellular accumulation of the glycosphingolipid GM2 ganglioside in KO but not WT mice. This cellular storage phenotype was most abundant in neurons of lamina I-II of the dorsal horn, a major relay site in the processing of painful stimuli. Spinal cords of KO mice also exhibited changes in astroglial and microglial populations throughout the gray matter suggestive of a neuroinflammatory process. Our findings establish the Slc9a6 KO mouse as a relevant tool for studying the sensory deficits in CS, and highlight selective vulnerabilities in relevant cell populations that may contribute to this phenotype. How NHE6 loss of function leads to such a multifaceted neurological syndrome is still undefined, and it is likely that NHE6 is involved with many cellular processes critical to normal nervous system development and function. In addition, the sensory issues exhibited by Slc9a6 KO mice, in combination with our neuropathological findings, are consistent with NHE6 loss of function impacting the entire nervous system. Sensory dysfunction in intellectually disabled individuals is challenging to assess and may impair patient safety and quality of life. Further mechanistic studies of the neurological impairments underlying CS and other genetic intellectual disability disorders must also take into account mechanisms affecting broader nervous system function in order to understand the full range of associated disabilities.


Asunto(s)
Ataxia/etiología , Endosomas/patología , Epilepsia/etiología , Enfermedades Genéticas Ligadas al Cromosoma X/etiología , Discapacidad Intelectual/etiología , Lisosomas/patología , Microcefalia/etiología , Trastornos de la Motilidad Ocular/etiología , Trastornos de la Sensación/etiología , Animales , Ataxia/genética , Ataxia/patología , Modelos Animales de Enfermedad , Epilepsia/genética , Epilepsia/patología , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Microcefalia/genética , Microcefalia/patología , Trastornos de la Motilidad Ocular/genética , Trastornos de la Motilidad Ocular/patología , Trastornos de la Sensación/genética , Trastornos de la Sensación/patología
10.
J Autoimmun ; 96: 59-73, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30174216

RESUMEN

Neuropsychiatric manifestations in lupus (NPSLE) affect ∼20-40% of patients. In the central nervous system, lipocalin-2 (LCN2) can promote injury through mechanisms directly linked to NPSLE, including brain barrier disruption, neurotoxicity, and glial activation. Since LCN2 is elevated in lupus and has been implicated in neuroinflammation, we investigated whether LCN2 is required for the pathogenesis of NPSLE. Here, we investigated the effects of LCN2 deficiency on the development of neurobehavioral deficits in the B6.Sle1.Sle3 (Sle1,3) mouse lupus model. Sle1,3 mice exhibited depression-like behavior and impaired spatial and recognition memory, and these deficits were attenuated in Sle1,3-LCN2KO mice. Whole-brain flow cytometry showed a significant increase in brain infiltrating leukocytes in Sle1,3 mice that was not reduced by LCN2 deficiency. RNA sequencing on sorted microglia revealed that several genes differentially expressed between B6 and Sle1,3 mice were regulated by LCN2, and that these genes are key mediators of the neuroinflammatory cascade. Importantly, LCN2 is upregulated in the cerebrospinal fluid of NPSLE patients across 2 different ethnicities. Our findings establish the Sle1,3 strain as an NPSLE model, demonstrate that LCN2 is a major regulator of the detrimental neuroimmune response in NPSLE, and identify CSF LCN2 as a novel biomarker for NPSLE.


Asunto(s)
Biomarcadores/metabolismo , Leucocitos/inmunología , Lipocalina 2/metabolismo , Vasculitis por Lupus del Sistema Nervioso Central/metabolismo , Inflamación Neurogénica/metabolismo , Animales , Barrera Hematoencefálica , Modelos Animales de Enfermedad , Femenino , Humanos , Lipocalina 2/antagonistas & inhibidores , Lipocalina 2/genética , Vasculitis por Lupus del Sistema Nervioso Central/diagnóstico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Inflamación Neurogénica/diagnóstico , Regulación hacia Arriba
11.
Front Immunol ; 9: 2189, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319641

RESUMEN

About 40% of patients with systemic lupus erythematosus experience diffuse neuropsychiatric manifestations, including impaired cognition and depression. Although the pathogenesis of diffuse neuropsychiatric SLE (NPSLE) is not fully understood, loss of brain barrier integrity, autoreactive antibodies, and pro-inflammatory cytokines are major contributors to disease development. Fingolimod, a sphingosine-1-phosphate (S1P) receptor modulator, prevents lymphocyte egress from lymphoid organs through functional antagonism of S1P receptors. In addition to reducing the circulation of autoreactive lymphocytes, fingolimod has direct neuroprotective effects such as preserving brain barrier integrity and decreasing pro-inflammatory cytokine secretion by astrocytes and microglia. Given these effects, we hypothesized that fingolimod would attenuate neurobehavioral deficits in MRL-lpr/lpr (MRL/lpr) mice, a validated neuropsychiatric lupus model. Fingolimod treatment was initiated after the onset of disease, and mice were assessed for alterations in cognitive function and emotionality. We found that fingolimod significantly attenuated spatial memory deficits and depression-like behavior in MRL/lpr mice. Immunofluorescent staining demonstrated a dramatic lessening of brain T cell and macrophage infiltration, and a significant reduction in cortical leakage of serum albumin, in fingolimod treated mice. Astrocytes and endothelial cells from treated mice exhibited reduced expression of inflammatory genes, while microglia showed differential regulation of key immune pathways. Notably, cytokine levels within the cortex and hippocampus were not appreciably decreased with fingolimod despite the improved neurobehavioral profile. Furthermore, despite a reduction in splenomegaly, lymphadenopathy, and circulating autoantibody titers, IgG deposition within the brain was unaffected by treatment. These findings suggest that fingolimod mediates attenuation of NPSLE through a mechanism that is not dependent on reduction of autoantibodies or cytokines, and highlight modulation of the S1P signaling pathway as a novel therapeutic target in lupus involving the central nervous system.


Asunto(s)
Depresión/inmunología , Clorhidrato de Fingolimod/farmacología , Vasculitis por Lupus del Sistema Nervioso Central/psicología , Lisofosfolípidos/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Animales , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Autoanticuerpos/inmunología , Técnicas de Observación Conductual , Conducta Animal/efectos de los fármacos , Encéfalo/citología , Encéfalo/inmunología , Encéfalo/fisiología , Cognición/efectos de los fármacos , Cognición/fisiología , Citocinas/inmunología , Depresión/tratamiento farmacológico , Depresión/psicología , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Vasculitis por Lupus del Sistema Nervioso Central/tratamiento farmacológico , Vasculitis por Lupus del Sistema Nervioso Central/genética , Vasculitis por Lupus del Sistema Nervioso Central/inmunología , Lisofosfolípidos/inmunología , Ratones , Ratones Endogámicos MRL lpr , Microglía/efectos de los fármacos , Microglía/inmunología , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/inmunología , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal/inmunología , Esfingosina/inmunología , Esfingosina/metabolismo , Resultado del Tratamiento
12.
Neuropharmacology ; 139: 76-84, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29990472

RESUMEN

Methotrexate is a dihydrofolate reductase inhibitor widely employed in curative treatment for children with acute lymphoblastic leukemia (ALL). However, methotrexate administration is also associated with persistent cognitive deficits among long-term childhood cancer survivors. Animal models of methotrexate-induced cognitive deficits have primarily utilized adult animals. The purpose of present study is to investigate the neurotoxicity of methotrexate in juvenile rats and its relevant mechanisms. The doses and schedule of systemic and intrathecal methotrexate, given from post-natal age 3-7 weeks, were chosen to model the effects of repeated methotrexate dosing on the developing brains of young children with ALL. This methotrexate regimen had no visible acute toxicity and no effect on growth. At 15 weeks of age (8 weeks after the last methotrexate dose) both spatial pattern memory and visual recognition memory were impaired. In addition, methotrexate-treated animals demonstrated impaired performance in the set-shifting assay, indicating decreased cognitive flexibility. Histopathological analysis demonstrated decreased cell proliferation in methotrexate-treated animals compared to controls, as well as changes in length and thickness of the corpus callosum. Moreover, methotrexate suppressed microglia activation and RANTES production. In conclusion, our study demonstrated that a clinically relevant regimen of systemic and intrathecal methotrexate induces persistent deficits in spatial pattern memory, visual recognition memory and executive function, lasting at least 8 weeks after the last injection. The mechanisms behind methotrexate-induced deficits are likely multifactorial and may relate to suppression of neurogenesis, alterations in neuroinflammation and microglial activation, and structural changes in the corpus callosum.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Trastornos del Conocimiento/inducido químicamente , Antagonistas del Ácido Fólico/efectos adversos , Trastornos de la Memoria/inducido químicamente , Metotrexato/efectos adversos , Animales , Encéfalo/patología , Proliferación Celular/efectos de los fármacos , Trastornos del Conocimiento/patología , Función Ejecutiva/efectos de los fármacos , Femenino , Antagonistas del Ácido Fólico/administración & dosificación , Homocisteína/análogos & derivados , Homocisteína/líquido cefalorraquídeo , Masculino , Trastornos de la Memoria/patología , Metotrexato/administración & dosificación , Microglía/efectos de los fármacos , Microglía/patología , Reconocimiento Visual de Modelos/efectos de los fármacos , Ratas Long-Evans , Reconocimiento en Psicología/efectos de los fármacos , Memoria Espacial/efectos de los fármacos
13.
J Pediatr Hematol Oncol ; 40(2): e91-e96, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28654460

RESUMEN

Liposomal cytarabine is currently being tested clinically as an alternative to intrathecal (IT) methotrexate (MTX) for preventing relapse within the central nervous system among patients with acute lymphoblastic leukemia. To compare the toxicity and cognitive deficits caused by IT MTX versus liposomal cytarabine, juvenile Long Evans rats were treated with IT injections of MTX 1 mg/kg×4 doses over 8 days, or liposomal cytarabine 0.8 mg once. Mean concentrations of free cytarabine in cerebrospinal fluid remained above the cytotoxic threshold of 0.4 µM for 2 weeks after dosing. Animals treated with liposomal cytarabine exhibited normal recognition and spatial memory 4 weeks after injection. In contrast, exposure to IT MTX led to impaired cognitive function. In addition, mean hematocrit on day 11 was significantly lower in the MTX-treated animals (30.8%; 95% confidence interval, 27.0%-34.7%; n=6) compared with that in the liposomal cytarabine-treated animals (39.5%; 95% confidence interval, 38.4%-40.6%; n=6; P<0.0001). Our data suggest that liposomal cytarabine induces fewer neurocognitive deficits and less acute hematologic toxicity compared with IT MTX. Liposomal cytarabine may therefore have therapeutic advantages over IT MTX, if it is equally effective in preventing relapse.


Asunto(s)
Antimetabolitos Antineoplásicos/toxicidad , Cognición/efectos de los fármacos , Citarabina/toxicidad , Hematopoyesis/efectos de los fármacos , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Citarabina/administración & dosificación , Preparaciones de Acción Retardada/toxicidad , Modelos Animales de Enfermedad , Femenino , Liposomas , Masculino , Ratas , Ratas Long-Evans
14.
Glia ; 65(12): 2051-2069, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28925029

RESUMEN

The TAM (Tyro3, Axl, and MerTK) family of receptor tyrosine kinases (RTKs) and their ligands, Gas6 and ProS1, are important for innate immune responses and central nervous system (CNS) homeostasis. While only Gas6 directly activates Axl, ProS1 activation of Tyro3/MerTK can indirectly activate Axl through receptor heterodimerization. Therefore, we generated Gas6-/- Axl-/- double knockout (DKO) mice to specifically examine the contribution of this signaling axis while retaining ProS1 signaling through Tyro3 and MerTK. We found that naïve young adult DKO and WT mice have comparable myelination and equal numbers of axons and oligodendrocytes in the corpus callosum. Using the cuprizone model of demyelination/remyelination, transmission electron microscopy revealed extensive axonal swellings containing autophagolysosomes and multivesicular bodies, and fewer myelinated axons in brains of DKO mice at 3-weeks recovery from a 6-week cuprizone diet. Analysis of immunofluorescent staining demonstrated more SMI32+ and APP+ axons and less myelin in the DKO mice. There were no significant differences in the number of GFAP+ astrocytes or Iba1+ microglia/macrophages between the groups of mice. However, at 6-weeks cuprizone and recovery, DKO mice had increased proinflammatory cytokine and altered suppressor of cytokine signaling (SOCS) mRNA expression supporting a role for Gas6-Axl signaling in proinflammatory cytokine suppression. Significant motor deficits in DKO mice relative to WT mice on cuprizone were also observed. These data suggest that Gas6-Axl signaling plays an important role in maintaining axonal integrity and regulating and reducing CNS inflammation that cannot be compensated for by ProS1/Tyro3/MerTK signaling.


Asunto(s)
Axones/patología , Regulación de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Trastornos del Movimiento , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Tirosina Quinasas Receptoras/deficiencia , Remielinización/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Axones/ultraestructura , Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Encefalitis/inducido químicamente , Encefalitis/patología , Regulación de la Expresión Génica/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibidores de la Monoaminooxidasa/toxicidad , Trastornos del Movimiento/etiología , Trastornos del Movimiento/genética , Trastornos del Movimiento/patología , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/patología , Vaina de Mielina/ultraestructura , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Propiocepción/efectos de los fármacos , Propiocepción/genética , Proteínas Proto-Oncogénicas/genética , Desempeño Psicomotor/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/genética , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/genética , Remielinización/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tirosina Quinasa del Receptor Axl
15.
J Neurochem ; 142(1): 118-131, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28407315

RESUMEN

The metabolism of tryptophan through kynurenine and serotonin pathways is linked to depression. Here, effects of different drugs with antidepressant properties (vortioxetine, fluoxetine, and ketamine) on various tryptophan metabolites in different brain regions and plasma were examined using tandem mass spectrometry (LC-MS/MS), in Flinders Sensitive Line rats, a genetic rat model of depression, and its controls: Flinders Sensitive Line and Sprague-Dawley rats. Protein levels of kynurenine pathway enzymes were measured in the brains and livers of these rat strains. Furthermore, effects of vortioxetine on tryptophan metabolites were assessed in the cortical regions of lupus mice (MRL/MpJ-FasIpr ), a murine model of increased depression-like behavior associated with inflammation. Sustained vortioxetine or fluoxetine (at doses aimed to fully occupy serotonin transporter via food or drinking water for at least 14 days) reduced levels of the excitotoxin quinolinic acid (QUIN) in various brain regions in all rats. Furthermore, chronic vortioxetine reduced levels of QUIN in MRL/MpJ-FasIpr mice. Acute i.p. administration of fluoxetine (10 mg/kg) or vortioxetine (10 mg/kg) led to reduced brain 5-hydroxyindoleacetic acid in Sprague-Dawley rats (2, 4, 6, and 8 h) and a similar trend was evident in Flinders Sensitive Line and Flinders Sensitive Line rats after 4 h. In contrast, single or repeated administration of ketamine (15 mg/kg i.p.) did not induce significant changes in metabolite levels. In conclusion, sustained vortioxetine and fluoxetine administration decreased QUIN independent of species, while ketamine was ineffective. These results support the hypothesis that modulating tryptophan metabolism may be part of the mechanism of action for some antidepressants.


Asunto(s)
Antidepresivos/farmacología , Depresión/metabolismo , Depresión/psicología , Triptófano/metabolismo , Animales , Química Encefálica/efectos de los fármacos , Fluoxetina/farmacología , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo , Ácido Hidroxiindolacético/metabolismo , Ketamina/farmacología , Quinurenina/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Masculino , Ratones , Piperazinas/farmacología , Ácido Quinolínico/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Sulfuros/farmacología , Vortioxetina
16.
Neuropharmacology ; 121: 89-99, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28414050

RESUMEN

Treatment-related sexual dysfunction is a common side effect of antidepressants and contributes to patient non-compliance or treatment cessation. However, the multimodal antidepressant, vortioxetine, demonstrates low sexual side effects in depressed patients. To investigate the mechanisms involved, sexual behavior was assessed in male and female rats after acute, and repeated (7 and 14 days) treatment with vortioxetine, flesinoxan (a 5-HT1A receptor agonist), CP-94253 (a 5-HT1B receptor agonist), or ondansetron (a 5-HT3 receptor antagonist). These selective ligands were chosen to simulate vortioxetine's direct modulation of these receptors. Paroxetine was also included in the male study. Acute and repeated treatment with vortioxetine at doses corresponding to clinical levels (based on serotonin transporter occupancy) had minimal effects on sexual behavior in male and female rats. High dose vortioxetine plus flesinoxan (to mimic predicted clinical levels of 5-HT1A receptor occupancy by vortioxetine) facilitated male rat sexual behavior (acutely) while inhibiting female rat proceptive behavior (both acutely and after 14 days treatment). The selective serotonin reuptake inhibitor, paroxetine, inhibited male sexual behavior after repeated administration (7 and 14 days). Flesinoxan alone facilitated male sexual behavior acutely while inhibiting female rat proceptive behavior after repeated administration (7 and 14 days). CP-94253 inhibited sexual behavior in both male and female rats after repeated administration. Ondansetron had no effect on sexual behavior. These findings underline the complex serotonergic regulation of sexual behavior and indicate that the low sexual side effects of vortioxetine found in clinical studies are likely associated with its direct modulation of serotonin receptors.


Asunto(s)
Piperazinas/farmacología , Receptores de Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Caracteres Sexuales , Conducta Sexual Animal/efectos de los fármacos , Sulfuros/farmacología , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Análisis de Varianza , Animales , Autorradiografía , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Proteínas de Unión al ARN/metabolismo , Ratas , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos , Agonistas de Receptores de Serotonina/farmacología , Factores de Tiempo , Vortioxetina
17.
Psychoneuroendocrinology ; 80: 92-98, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28324704

RESUMEN

Depression and anxiety are the most common psychiatric disorders, representing a major public health concern. Dysregulation of oxidative and inflammatory systems may be associated with psychiatric disorders, such as depression and anxiety. Due to the need to find appropriate animal models to the understanding of such disorders, we queried whether 2 BXD recombinant inbred (RI) mice strains (BXD21/TyJ RI and BXD84/RwwJ RI mice) and C57BL/6 wild-type mice show differential performance in depression and anxiety related behaviors and biomarkers. Specifically, we assessed social preference, elevated plus maze, forced swim, and Von Frey tests at 3-4 months-of-age, as well as activation of cytokines and antioxidant mRNA levels in the cortex at 7 months-of-age. We report that (1) the BXD84/RwwJ RI strain exhibits anxiety disorder and social avoidance-like behavior (2) BXD21/TyJ RI strain shows a resistance to depression illness, and (3) sex-dependent cytokine profiles and allodynia with elevated inflammatory activity were inherent to male BXD21/TyJ RI mice. In conclusion, we provide novel data in favor of the use of BXD recombinant inbred mice to further understand anxiety and depression disorders.


Asunto(s)
Trastornos de Ansiedad/metabolismo , Trastorno Depresivo/metabolismo , Trastorno de la Conducta Social/genética , Animales , Ansiedad/genética , Ansiedad/metabolismo , Trastornos de Ansiedad/genética , Conducta Animal/fisiología , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Depresión/genética , Depresión/metabolismo , Trastorno Depresivo/genética , Modelos Animales de Enfermedad , Femenino , Hiperalgesia/genética , Hiperalgesia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Caracteres Sexuales , Conducta Social , Trastorno de la Conducta Social/metabolismo
18.
Int J Neuropsychopharmacol ; 20(6): 510-515, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28158336

RESUMEN

Background: Cognitive dysfunction is among the key symptoms of major depressive disorder and can be affected by antidepressants. Cognitive decline also occurs in normal aging. The effects of different antidepressants on affective and cognitive domains in older subjects are seldom assessed simultaneously. Methods: Healthy middle-aged female mice received vehicle or antidepressant (vortioxetine, vilazodone, duloxetine, or fluoxetine) at therapeutic doses. After 1 month treatment, mice were accessed for visuospatial memory and depression-like behavior. A separate cohort of mice received 3 months of treatment and was test for recognition memory and depression-like behavior. Results: After 1 month treatment, vortioxetine improved visuospatial memory and reduced depression-like behavior. Vilazodone reduced depression-like behavior. Duloxetine and fluoxetine were ineffective in both tests. After 3 months treatment, vortioxetine reduced depression-like behavior without affecting recognition memory, while fluoxetine impaired recognition memory. Duloxetine and vilazodone had no effect in both tests. Conclusion: Different antidepressants have distinct effects in middle-aged female mice.


Asunto(s)
Envejecimiento/efectos de los fármacos , Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Memoria/efectos de los fármacos , Psicotrópicos/farmacología , Envejecimiento/psicología , Animales , Cognición/efectos de los fármacos , Estudios de Cohortes , Clorhidrato de Duloxetina/farmacología , Femenino , Fluoxetina/farmacología , Ratones Endogámicos C57BL , Piperazinas/farmacología , Distribución Aleatoria , Percepción Espacial/efectos de los fármacos , Sulfuros/farmacología , Clorhidrato de Vilazodona/farmacología , Percepción Visual/efectos de los fármacos , Vortioxetina
19.
J Psychopharmacol ; 31(3): 365-376, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27678087

RESUMEN

Neuroplasticity is fundamental for brain functions, abnormal changes of which are associated with mood disorders and cognitive impairment. Neuroplasticity can be affected by neuroactive medications and by aging. Vortioxetine, a multimodal antidepressant, has shown positive effects on cognitive functions in both pre-clinical and clinical studies. In rodent studies, vortioxetine increases glutamate neurotransmission, promotes dendritic branching and spine maturation, and elevates hippocampal expression of the activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) at the transcript level. The present study aims to assess the effects of vortioxetine on several neuroplasticity-related molecules in different experimental systems. Chronic (1 month) vortioxetine increased Arc/Arg3.1 protein levels in the cortical synaptosomes of young and middle-aged mice. In young mice, this was accompanied by an increase in actin-depolymerizing factor (ADF)/cofilin serine 3 phosphorylation without altering the total ADF/cofilin protein level, and an increase in the GluA1 subunit of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor phosphorylation at serine 845 (S845) without altering serine 831 (S831) GluA1 phosphorylation nor the total GluA1 protein level. Similar effects were detected in cultured rat hippocampal neurons: Acute vortioxetine increased S845 GluA1 phosphorylation without changing S831 GluA1 phosphorylation or the total GluA1 protein level. These changes were accompanied by an increase in α subunit of Ca2+/calmodulin-dependent kinase (CaMKIIα) phosphorylation (at threonine 286) without changing the total CaMKIIα protein level in cultured neurons. In addition, chronic (1 month) vortioxetine, but not fluoxetine, restored the age-associated reduction in Arc/Arg3.1 and c-Fos transcripts in the frontal cortex of middle-aged mice. Taken together, these results demonstrated that vortioxetine modulates molecular targets that are related to neuroplasticity.


Asunto(s)
Plasticidad Neuronal/efectos de los fármacos , Piperazinas/farmacología , Sulfuros/farmacología , Animales , Antidepresivos/farmacología , Proteínas del Citoesqueleto/metabolismo , Femenino , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Serina/metabolismo , Vortioxetina
20.
Neurobiol Dis ; 96: 144-155, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27623015

RESUMEN

The mutation in huntingtin (mHtt) leads to a spectrum of impairments in the developing forebrain of Huntington's disease (HD) mouse models. Whether these developmental alterations are due to loss- or gain-of-function mechanisms and contribute to HD pathogenesis is unknown. We examined the role of selective loss of huntingtin (Htt) function during development on postnatal vulnerability to cell death. We employed mice expressing very low levels of Htt throughout embryonic life to postnatal day 21 (Hdhd•hyp). We demonstrated that Hdhd•hyp mice exhibit: (1) late-life striatal and cortical neuronal degeneration; (2) neurological and skeletal muscle alterations; and (3) white matter tract impairments and axonal degeneration. Hdhd•hyp embryos also exhibited subpallial heterotopias, aberrant striatal maturation and deregulation of gliogenesis. These results indicate that developmental deficits associated with Htt functions render cells present at discrete neural foci increasingly susceptible to cell death, thus implying the potential existence of a loss-of-function developmental component to HD pathogenesis.


Asunto(s)
Discapacidades del Desarrollo/genética , Proteína Huntingtina/deficiencia , Enfermedad de Huntington/complicaciones , Enfermedad de Huntington/genética , Mutación/genética , Enfermedades Neurodegenerativas/etiología , Factores de Edad , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Discapacidades del Desarrollo/complicaciones , Modelos Animales de Enfermedad , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Proteína Huntingtina/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/complicaciones , Trastornos Psicomotores/etiología , Trastornos Psicomotores/genética , ARN Mensajero/metabolismo , Sustancia Blanca/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...