Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(51): 25982-25990, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31792184

RESUMEN

Retrotransposons compose a staggering 40% of the mammalian genome. Among them, endogenous retroviruses (ERV) represent sequences that closely resemble the proviruses created from exogenous retroviral infection. ERVs make up 8 to 10% of human and mouse genomes and range from evolutionarily ancient sequences to recent acquisitions. Studies in Drosophila have provided a causal link between genomic retroviral elements and cognitive decline; however, in mammals, the role of ERVs in learning and memory remains unclear. Here we studied 2 independent murine models for ERV activation: muMT strain (lacking B cells and antibody production) and intracerebroventricular injection of streptozotocin (ICVI-STZ). We conducted behavioral assessments (contextual fear memory and spatial learning), as well as gene and protein analysis (RNA sequencing, PCR, immunohistochemistry, and western blot assays). Mice lacking mitochondrial antiviral-signaling protein (MAVS) and mice lacking stimulator of IFN genes protein (STING), 2 downstream sensors of ERV activation, provided confirmation of ERV impact. We found that muMT mice and ICVI-STZ mice induced hippocampal ERV activation, as shown by increased gene and protein expression of the Gag sequence of the transposable element intracisternal A-particle. ERV activation was accompanied by significant hippocampus-related memory impairment in both models. Notably, the deficiency of the MAVS pathway was protective against ICVI-STZ-induced cognitive pathology. Overall, our results demonstrate that ERV activation is associated with cognitive impairment in mice. Moreover, they provide a molecular target for strategies aimed at attenuating retroviral element sensing, via MAVS, to treat dementia and neuropsychiatric disorders.


Asunto(s)
Retrovirus Endógenos/genética , Hipocampo/virología , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/virología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Conducta Animal , Encéfalo/patología , Disfunción Cognitiva , Elementos Transponibles de ADN , Modelos Animales de Enfermedad , Retrovirus Endógenos/fisiología , Regulación de la Expresión Génica , Productos del Gen gag , Hipocampo/efectos de los fármacos , Aprendizaje , Masculino , Proteínas de la Membrana/metabolismo , Memoria , Trastornos de la Memoria/psicología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estreptozocina/farmacología
2.
Mol Med ; 27(1): 99, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34488619

RESUMEN

BACKGROUND: We have found disruption of expression of major transcriptional regulators of circadian rhythm in the kidneys of several mouse models of lupus nephritis. Here we define the consequence of this disturbance with respect to circadian gene expression and renal homeostatic function in a mouse model of lupus nephritis. METHODS: Molecular profiling of kidneys from 47 young and 41 nephritic female NZB/W F1 mice was performed at 4 hourly intervals over a 24 h period. Disruption of major circadian transcriptional regulators was confirmed by qPCR. Molecular data was normalized and analyzed for rhythmicity using RAIN analysis. Serum aldosterone and glucose and urine sodium and potassium were measured at 4 hourly intervals in pre-nephritic and nephritic mice and blood pressure was measured every 4 h. Analyses were repeated after induction of complete remission of nephritis using combination cyclophosphamide and costimulatory blockade. RESULTS: We show a profound alteration of renal circadian rhythms in mice with lupus nephritis affecting multiple renal pathways. Using Cosinor analysis we identified consequent alterations of renal homeostasis and metabolism as well as blood pressure dipper status. This circadian dysregulation was partially reversed by remission induction therapy. CONCLUSIONS: Our studies indicate the role of inflammation in causing the circadian disruption and suggest that screening for loss of normal blood pressure dipping should be incorporated into LN management. The data also suggest a potential role for circadian agonists in the treatment of lupus nephritis.


Asunto(s)
Biomarcadores , Ritmo Circadiano/genética , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Nefritis Lúpica/etiología , Nefritis Lúpica/metabolismo , Animales , Biología Computacional/métodos , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Nefritis Lúpica/patología , Ratones , Transcriptoma
3.
Mol Med ; 26(1): 34, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32303185

RESUMEN

BACKGROUND: Macrophage Migration Inhibitory Factor (MIF) is a potent proinflammatory cytokine that promotes the production of other immune mediators. MIF is produced by most cell types in the brain including microglia, astrocytes and neurons. Enhanced expression of MIF might contribute to the persistent activation of glial, chronic neuroinflammation and neurodegeneration. Here, we investigated the effect of MIF on inflammatory markers and spatial learning in a mouse model of sporadic AD and on tau pathology in AD patients. METHODS: We examined the effects of MIF deficiency and pharmacological MIF inhibition in vitro and in vivo. In vitro, quantitative PCR and ELISA were used to assess cytokine production of STZ-treated glial cells. In vivo, C57BL/6 mice were subjected to intracerebroventricular streptozotocin injection (3 mg/kg, ICV-STZ). Neuroinflammation and contextual learning performance were assessed using quantitative PCR and fear conditioning, respectively. Pharmacological MIF inhibition was achieved with intraperitoneal injections of ISO-1 (daily, IP, 20 mg/kg in 5% DMSO in 0.9% NaCl) for 4 weeks following ICV-STZ injection. The findings from ISO-1 treated mice were confirmed in MIF knockout C57BL/6. To assess the role of MIF in human AD, cerebrospinal fluid levels of MIF and hyperphosphorylated tau were measured using ELISA. RESULTS: Administration ICV-STZ resulted in hippocampal dependent cognitive impairment. MIF inhibition with ISO-1 significantly improved the STZ-induced impairment in contextual memory performance, indicating MIF-related inflammation as a major contributor to ICV-STZ-induced memory deficits. Furthermore, inhibition of the MIF resulted in reduced cytokine production in vitro and in vivo. In human subjects with AD at early clinical stages, cerebrospinal fluid levels of MIF were increased in comparison with age-matched controls, and correlated with biomarkers of tau hyper-phosphorylation and neuronal injury hinting at MIF levels as a potential biomarker for early-stage AD. CONCLUSIONS: The present study indicates the key role of MIF in controlling the chronic cytokine release in neuroinflammation related to tau hyperphosphorylation, neurodegeneration, and clinical manifestations of AD, suggesting the potential of MIF inhibition as therapeutic strategy to slow down neurodegeneration and clinical disease progression.


Asunto(s)
Enfermedad de Alzheimer/etiología , Disfunción Cognitiva/genética , Inflamación/genética , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Degeneración Nerviosa/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Animales , Astrocitos/metabolismo , Biomarcadores , Células Cultivadas , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/psicología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Espacio Extracelular/metabolismo , Femenino , Regulación de la Expresión Génica , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Masculino , Memoria/efectos de los fármacos , Ratones , Microglía/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología
4.
Mol Med ; 25(1): 24, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31146675

RESUMEN

BACKGROUND: Alcohol abuse affects the brain regions responsible for memory, coordination and emotional processing. Binge alcohol drinking has shown reductions in brain activity, but the molecular targets have not been completely elucidated. We hypothesized that brain cells respond to excessive alcohol by releasing a novel inflammatory mediator, called cold inducible RNA-binding protein (CIRP), which is critical for the decreased brain metabolic activity and impaired cognition. METHODS: Male wild type (WT) mice and mice deficient in CIRP (CIRP-/-) were studied before and after exposure to binge alcohol level by assessment of relative brain glucose metabolism with fluorodeoxyglucose (18FDG) and positron emission tomography (PET). Mice were also examined for object-place memory (OPM) and open field (OF) tasks. RESULTS: Statistical Parametric Analysis (SPM) of 18FDG-PET uptake revealed marked decreases in relative glucose metabolism in distinct brain regions of WT mice after binge alcohol. Regional analysis (post hoc) revealed that while activity in the temporal (secondary visual) and limbic (entorhinal/perirhinal) cortices was decreased in WT mice, relative glucose metabolic activity was less suppressed in the CIRP-/- mice. Group and condition interaction analysis revealed differing responses in relative glucose metabolism (decrease in WT mice but increase in CIRP-/- mice) after alcohol in brain regions including the hippocampus and the cortical amygdala where the percent changes in metabolic activity correlated with changes in object discrimination performance. Behaviorally, alcohol-treated WT mice were impaired in exploring a repositioned object in the OPM task, and were more anxious in the OF task, whereas CIRP-/- mice were not impaired in these tasks. CONCLUSION: CIRP released from brain cells could be responsible for regional brain metabolic hypoactivity leading to cognitive impairment under binge alcohol conditions.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/complicaciones , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Proteínas de Unión al ARN/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Fluorodesoxiglucosa F18/análisis , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Tomografía de Emisión de Positrones , Proteínas de Unión al ARN/genética , Memoria Espacial/efectos de los fármacos
5.
Mol Med ; 22: 789-799, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27878209

RESUMEN

Long-term sepsis survivors sustain cryptic brain injury that leads to cognitive impairment, emotional imbalance, and increased disability burden. Suitable animal models of sepsis, such as cecal ligation and puncture (CLP), have permitted the analysis of abnormal brain circuits that underlie post-septic behavioral phenotypes. For instance, we have previously shown that CLP-exposed mice exhibit impaired spatial memory together with depleted dendritic arbors and decreased spines in the apical dendrites of pyramidal neurons in the CA1 region of the hippocampus. Here we show that contextual fear conditioning, a form of associative memory for fear, is chronically disrupted in CLP mice when compared to SHAM-operated animals. We also find that the excitatory neurons in the basolateral nucleus of the amygdala (BLA) and the granule cells in the dentate gyrus (DG) display significantly fewer dendritic spines in the CLP group relative to the SHAM mice, although the dendritic arbors and gross morphology of the BLA and DG are comparable between the two groups. Moreover, the basal dendrites of CA1 pyramidal neurons are unaffected in the CLP mice. Taken together, our data indicate that the structural damage in the amygdalar-hippocampal network represents the neural substrate for impaired contextual fear memory in long-term sepsis survivors. Further, our data suggest that the brain injury caused by overwhelming sepsis alters the stability of the synaptic connections involved in associative fear. These results likely have implications for the emotional imbalance observed in human sepsis survivors.

6.
bioRxiv ; 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38405863

RESUMEN

Aims: This study aims to elucidate the underlying mechanisms of diving reflex, a powerful endogenous mechanism supporting underwater mammalian survival. Antioxidative responses, observed in marine mammals, may be contributing factors. Using a multi-organ approach, this study assesses whether acute and chronic diving reflex activate nuclear factor-erythroid-2-related factor 2 (NRF2) signaling pathways, which regulate cellular antioxidant responses. Methods: Male Sprague-Dawley rats ( n =38) underwent either a single diving session to elicit acute diving reflex, or daily diving sessions for 4-weeks to produce chronic diving reflex. NRF2 (total, nuclear, phosphorylated), NRF2-downstream genes, and malondialdehyde were assessed via Western blot, immunofluorescence, RT-PCR, and ELISA in brain, lung, kidney, and serum. Results: Diving reflex increased nuclear NRF2, phosphorylated NRF2, and antioxidative gene expression, in an organ-specific and exposure time-specific manner. Comparing organs, the brain had the highest increase of phosphorylated NRF2 expression, while kidney had the highest degree of nuclear NRF2 expression. Comparing acute and chronic sessions, phosphorylated NRF2 increased the most with chronic diving reflex, but acute diving reflex had the highest antioxidative gene expression. Notably, calcitonin gene-related peptide appears to mediate diving reflex' effects on NRF2 activation. Conclusions: Acute and chronic diving reflex activate potent NRF2 signaling in the brain and peripheral organs. Interestingly, acute diving reflex induces higher expression of downstream antioxidative genes compared to chronic diving reflex. This result contradicts previous assumptions requiring chronic exposure to diving for induction of antioxidative effects and implies that the diving reflex has a strong translational potential during preconditioning and postconditioning therapies. Key Points: Diving reflex activates potent NRF2 signaling via multiple mechanisms, including phosphorylation, nuclear translocation, and KEAP1 downregulation with both acute and chronic exposure.Diving reflex activates NRF2 via differential pathways in the brain and other organs; phosphorylated NRF2 increases more in the brain, while nuclear NRF2 increases more in the peripheral organs.Acute diving reflex exposure induces a more pronounced antioxidative effect than chronic diving reflex exposure, indicating that the antioxidative response activated by diving reflex is not dependent upon chronic adaptive responses and supports diving reflex as both a preconditioning and postconditioning treatment.

7.
Front Neurosci ; 18: 1237748, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38384483

RESUMEN

Rodents establish dominance hierarchy as a social ranking system in which one subject acts as dominant over all the other subordinate individuals. Dominance hierarchy regulates food access and mating opportunities, but little is known about its significance in other social behaviors, for instance during collective navigation for foraging or migration. Here, we implemented a simplified goal-directed spatial task in mice, in which animals navigated individually or collectively with their littermates foraging for food. We compared between conditions and found that the social condition exerts significant influence on individual displacement patterns, even when efficient navigation rules leading to reward had been previously learned. Thus, movement patterns and consequent task performance were strongly dependent on contingent social interactions arising during collective displacement, yet their influence on individual behavior was determined by dominance hierarchy. Dominant animals did not behave as leaders during collective displacement; conversely, they were most sensitive to the social environment adjusting their performance accordingly. Social ranking in turn was associated with specific spontaneous neural activity patterns in the prefrontal cortex and hippocampus, with dominant mice showing higher firing rates, larger ripple oscillations, and stronger neuronal entrainment by ripples than subordinate animals. Moreover, dominant animals selectively increased their cortical spiking activity during collective movement, while subordinate mice did not modify their firing rates, consistent with dominant animals being more sensitive to the social context. These results suggest that dominance hierarchy influences behavioral performance during contingent social interactions, likely supported by the coordinated activity in the hippocampal-prefrontal circuit.

8.
Bioessays ; 33(8): 588-91, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21681774

RESUMEN

A new study entitled "Normal gut microbiota modulates brain development and behavior", published in the Proceedings of the National Academy of Sciences, requires that we reconsider the notion that the brain is an immune-privileged site. The authors demonstrate that intestinal microbiota must be present within a set time-frame for normal synaptogenesis to occur in the brain. In the absence of intestinal microbiota, histopathological and behavioral abnormalities arise. These observations necessitate a new look at the many interconnections of the immune system and the brain, suggesting new frontiers for research and new therapeutic strategies for neurodevelopmental diseases.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Factores Inmunológicos/metabolismo , Intestinos/microbiología , Neuroinmunomodulación , Animales , Conducta/fisiología , Encéfalo/inmunología , Encéfalo/fisiología , Citocinas/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Intestinos/inmunología , Potenciación a Largo Plazo , Metagenoma , Receptores Inmunológicos/metabolismo , Sinapsis/inmunología , Sinapsis/fisiología , Transmisión Sináptica
9.
Proc Natl Acad Sci U S A ; 107(43): 18569-74, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20921396

RESUMEN

Damaging interactions between antibodies and brain antigenic targets may be responsible for an expanding range of neurological disorders. In the case of systemic lupus erythematosus (SLE), patients generate autoantibodies (AAbs) that frequently bind dsDNA. Although some symptoms of SLE may arise from direct reactivity to dsDNA, much of the AAb-mediated damage originates from cross-reactivity with other antigens. We have studied lupus AAbs that bind dsDNA and cross-react with the NR2A and NR2B subunits of the NMDA receptor (NMDAR). In adult mouse models, when the blood-brain barrier is compromised, these NMDAR-reactive AAbs access the brain and elicit neuronal death with ensuing cognitive dysfunction and emotional disturbance. The cellular mechanisms that underlie these deleterious effects remain incompletely understood. Here, we show that, at low concentration, the NMDAR-reactive AAbs are positive modulators of receptor function that increase the size of NMDAR-mediated excitatory postsynaptic potentials, whereas at high concentration, the AAbs promote excitotoxicity through enhanced mitochondrial permeability transition. Other synaptic receptors are completely unaffected by the AAbs. NMDAR activation is required for producing both the synaptic and the mitochondrial effects. Our study thus reveals the mechanisms by which NMDAR-reactive AAbs trigger graded cellular alterations, which are likely to be responsible for the transient and permanent neuropsychiatric symptoms observed in patients with SLE. Our study also provides a model in which local AAb concentration determines the exact nature of the cellular response.


Asunto(s)
Autoanticuerpos/toxicidad , Encéfalo/inmunología , Encéfalo/fisiopatología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/fisiopatología , Vasculitis por Lupus del Sistema Nervioso Central/inmunología , Vasculitis por Lupus del Sistema Nervioso Central/fisiopatología , Neurotoxinas/toxicidad , Animales , Reacciones Cruzadas , Potenciales Postsinápticos Excitadores , Femenino , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Poro de Transición de la Permeabilidad Mitocondrial , Modelos Inmunológicos , Modelos Neurológicos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/inmunología
10.
Transl Stroke Res ; 2023 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-37493939

RESUMEN

Subarachnoid hemorrhage (SAH) is a severe form of stroke that can cause unpredictable and diffuse cerebral damage, which is difficult to detect until it becomes irreversible. Therefore, there is a need for a reliable method to identify dysfunctional regions and initiate treatment before permanent damage occurs. Neurobehavioral assessments have been suggested as a possible tool to detect and approximately localize dysfunctional cerebral regions. In this study, we hypothesized that a neurobehavioral assessment battery could be a sensitive and specific method for detecting damage in discrete cerebral regions following SAH. To test this hypothesis, a behavioral battery was employed at multiple time points after SAH induced via an endovascular perforation, and brain damage was confirmed via postmortem histopathological analysis. Our results demonstrate that impairment of sensorimotor function accurately predict damage in the cerebral cortex (AUC 0.905; sensitivity 81.8%; specificity 90.9%) and striatum (AUC 0.913; sensitivity 90.1%; specificity 100%), while impaired novel object recognition is a more accurate indicator of damage to the hippocampus (AUC 0.902; sensitivity 74.1%; specificity 83.3%) than impaired reference memory (AUC 0.746; sensitivity 72.2%; specificity 58.0%). Tests for anxiety-like and depression-like behaviors predict damage to the amygdala (AUC 0.900; sensitivity 77.0%; specificity 81.7%) and thalamus (AUC 0.963; sensitivity 86.3%; specificity 87.8%), respectively. This study suggests that recurring behavioral testing can accurately predict damage in specific brain regions, which could be developed into a clinical battery for early detection of SAH damage in humans, potentially improving early treatment and outcomes.

11.
Res Sq ; 2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37292945

RESUMEN

Subarachnoid hemorrhage (SAH) is a severe form of stroke that can cause unpredictable and diffuse cerebral damage, which is difficult to detect until it becomes irreversible. Therefore, there is a need for a reliable method to identify dysfunctional regions and initiate treatment before permanent damage occurs. Neurobehavioral assessments have been suggested as a possible tool to detect and approximately localize dysfunctional cerebral regions. In this study, we hypothesized that a neurobehavioral assessment battery could be a sensitive and specific early warning for damage in discrete cerebral regions following SAH. To test this hypothesis, a behavioral battery was employed at multiple time points after SAH induced via an endovascular perforation, and brain damage was confirmed via postmortem histopathological analysis. Our results demonstrate that impairment of sensorimotor function accurately predict damage in the cerebral cortex (AUC: 0.905; sensitivity: 81.8%; specificity: 90.9%) and striatum (AUC: 0.913; sensitivity: 90.1%; specificity: 100%), while impaired novel object recognition is a more accurate indicator of damage to the hippocampus (AUC: 0.902; sensitivity: 74.1%; specificity: 83.3%) than impaired reference memory (AUC: 0.746; sensitivity: 72.2%; specificity: 58.0%). Tests for anxiety-like and depression-like behaviors predict damage to the amygdala (AUC: 0.900; sensitivity: 77.0%; specificity: 81.7%) and thalamus (AUC: 0.963; sensitivity: 86.3%; specificity: 87.8%), respectively. This study suggests that recurring behavioral testing can accurately predict damage in specific brain regions, which could be developed into a clinical battery for early detection of SAH damage in humans, potentially improving early treatment and outcomes.

12.
Mol Med ; 18: 930-7, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22634723

RESUMEN

Severe sepsis, a syndrome that complicates infection and injury, affects 750,000 annually in the United States. The acute mortality rate is approximately 30%, but, strikingly, sepsis survivors have a significant disability burden: up to 25% of survivors are cognitively and physically impaired. To investigate the mechanisms underlying persistent cognitive impairment in sepsis survivors, here we developed a murine model of severe sepsis survivors following cecal ligation and puncture (CLP) to study cognitive impairments. We observed that serum levels of high mobility group box 1 (HMGB1), a critical mediator of acute sepsis pathophysiology, are increased in sepsis survivors. Significantly, these levels remain elevated for at least 4 wks after CLP. Sepsis survivors develop significant, persistent impairments in learning and memory, and anatomic changes in the hippocampus associated with a loss of synaptic plasticity. Administration of neutralizing anti-HMGB1 antibody to survivors, beginning 1 wk after onset of peritonitis, significantly improved memory impairments and brain pathology. Administration of recombinant HMGB1 to naïve mice recapitulated the memory impairments. Together, these findings indicate that elevated HMGB1 levels mediate cognitive decline in sepsis survivors, and suggest that it may be possible to prevent or reverse cognitive impairments in sepsis survivors by administration of anti-HMGB1 antibodies.


Asunto(s)
Trastornos del Conocimiento/sangre , Trastornos del Conocimiento/complicaciones , Proteína HMGB1/sangre , Sepsis/sangre , Sepsis/complicaciones , Animales , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/farmacología , Conducta Animal/efectos de los fármacos , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Proteína HMGB1/inmunología , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipocampo/fisiopatología , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Sepsis/fisiopatología , Análisis de Supervivencia
13.
Sci Transl Med ; 14(641): eabe9726, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35442708

RESUMEN

The fetal brain is constantly exposed to maternal IgG before the formation of an effective blood-brain barrier (BBB). Here, we studied the consequences of fetal brain exposure to an antibody to the astrocytic protein aquaporin-4 (AQP4-IgG) in mice. AQP4-IgG was cloned from a patient with neuromyelitis optica spectrum disorder (NMOSD), an autoimmune disease that can affect women of childbearing age. We found that embryonic radial glia cells in neocortex express AQP4. These cells are critical for blood vessel and BBB formation through modulation of the WNT signaling pathway. Male fetuses exposed to AQP4-IgG had abnormal cortical vasculature and lower expression of WNT signaling molecules Wnt5a and Wnt7a. Positron emission tomography of adult male mice exposed in utero to AQP4-IgG revealed increased blood flow and BBB leakiness in the entorhinal cortex. Adult male mice exposed in utero to AQP4-IgG had abnormal cortical vessels, fewer dendritic spines in pyramidal and stellate neurons, and more S100ß+ astrocytes in the entorhinal cortex. Behaviorally, they showed impairments in the object-place memory task. Neural recordings indicated that their grid cell system, within the medial entorhinal cortex, did not map the local environment appropriately. Collectively, these data implicate in utero binding of AQP4-IgG to radial glia cells as a mechanism for alterations of the developing male brain and adds NMOSD to the conditions in which maternal IgG may cause persistent brain dysfunction in offspring.


Asunto(s)
Autoanticuerpos , Neuromielitis Óptica , Animales , Acuaporina 4/metabolismo , Barrera Hematoencefálica/metabolismo , Femenino , Humanos , Inmunoglobulina G , Masculino , Ratones
14.
Mol Med ; 17(11-12): 1349-56, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21953419

RESUMEN

The mediators of tissue damage in systemic lupus erythematosus (SLE) such as antibodies, cytokines and activated immune cells have direct access to most organs in the body but must penetrate the blood-brain barrier (BBB) to gain access to brain tissue. We hypothesized that compromise of the BBB occurs episodically such that the brain will acquire tissue damage slowly and not at the same rate as other organs. On the basis of these assumptions, we wished to determine if duration of disease correlated with brain injury, as measured with functional magnetic resonance imaging (fMRI), and if this was independent of degree of tissue damage in other organs. We investigated differences in brain activation patterns using fMRI in 13 SLE patients stratified by disease duration of ≤2 years (short-term [ST]) or ≥10 years (long-term [LT]). Two fMRI paradigms were selected to measure working memory and emotional response (fearful faces task). Performance in the working memory task was significantly better in the ST group for one and two shape recall; however, both groups did poorly with three shape recall. Imaging studies demonstrated significantly increased cortical activation in the ST group in regions associated with cognition during the two shape retention phase of the working memory task (P < 0.001) and increased amygdala (P < 0.05) and superior parietal (P < 0.01) activation in response to the fearful faces paradigm. In conclusion, analysis of activation patterns stratified by performance accuracy, differences in co-morbid disease, corticosteroid doses or disease activity suggests that these observed differences are attributable to SLE effects on the central nervous system exclusive of vascular disease or other confounding influences. Our hypothesis is further supported by the lack of correlation between regional brain abnormalities on fMRI and the Systemic Lupus International Collaborating Clinics (SLICC) damage index.


Asunto(s)
Mapeo Encefálico , Encéfalo/patología , Encéfalo/fisiopatología , Lupus Eritematoso Sistémico/patología , Lupus Eritematoso Sistémico/fisiopatología , Imagen por Resonancia Magnética/métodos , Adulto , Cara , Miedo , Femenino , Humanos , Masculino , Memoria a Corto Plazo , Persona de Mediana Edad , Especificidad de Órganos , Análisis y Desempeño de Tareas , Factores de Tiempo , Adulto Joven
15.
Front Neurol ; 12: 721108, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721260

RESUMEN

Autism Spectrum Disorder (ASD) is a group of neurodevelopmental conditions that is four times more commonly diagnosed in males than females. While susceptibility genes located in the sex chromosomes have been identified in ASD, it is unclear whether they are sufficient to explain the male bias or whether gonadal hormones also play a key role. We evaluated the sex chromosomal and hormonal influences on the male bias in a murine model of ASD, in which mice are exposed in utero to a maternal antibody reactive to contactin-associated protein-like 2 (Caspr2), which was originally cloned from a mother of a child with ASD (termed C6 mice henceforth). In this model, only male mice are affected. We used the four-core-genotypes (FCG) model in which the Sry gene is deleted from the Y chromosome (Y-) and inserted into autosome 3 (TgSry). Thus, by combining the C6 and FCG models, we were able to differentiate the contributions of sex chromosomes and gonadal hormones to the development of fetal brain and adult behavioral phenotypes. We show that the presence of the Y chromosome, or lack of two X chromosomes, irrespective of gonadal sex, increased the susceptibility to C6-induced phenotypes including the abnormal growth of the developing fetal cerebral cortex, as well as a behavioral pattern of decreased open-field exploration in adult mice. Our results indicate that sex chromosomes are the main determinant of the male bias in the maternal C6-induced model of ASD. The less dominant hormonal effect may be due to modulation by sex chromosome genes of factors involved in gonadal hormone pathways in the brain.

16.
Bioelectron Med ; 7(1): 17, 2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34809706

RESUMEN

BACKGROUND: Extracellular recording represents a crucial electrophysiological technique in neuroscience for studying the activity of single neurons and neuronal populations. The electrodes capture voltage traces that, with the help of analytical tools, reveal action potentials ('spikes') as well as local field potentials. The process of spike sorting is used for the extraction of action potentials generated by individual neurons. Until recently, spike sorting was performed with manual techniques, which are laborious and unreliable due to inherent operator bias. As neuroscientists add multiple electrodes to their probes, the high-density devices can record hundreds to thousands of neurons simultaneously, making the manual spike sorting process increasingly difficult. The advent of automated spike sorting software has offered a compelling solution to this issue and, in this study, we present a simple-to-execute framework for running an automated spike sorter. METHODS: Tetrode recordings of freely-moving mice are obtained from the CA1 region of the hippocampus as they navigate a linear track. Tetrode recordings are also acquired from the prelimbic cortex, a region of the medial prefrontal cortex, while the mice are tested in a T maze. All animals are implanted with custom-designed, 3D-printed microdrives that carry 16 electrodes, which are bundled in a 4-tetrode geometry. RESULTS: We provide an overview of a framework for analyzing single-unit data in which we have concatenated the acquisition system (Cheetah, Neuralynx) with analytical software (MATLAB) and an automated spike sorting pipeline (MountainSort). We give precise instructions on how to implement the different steps of the framework, as well as explanations of our design logic. We validate this framework by comparing manually-sorted spikes against automatically-sorted spikes, using neural recordings of the hippocampus and prelimbic cortex in freely-moving mice. CONCLUSIONS: We have efficiently integrated the MountainSort spike sorter with Neuralynx-acquired neural recordings. Our framework is easy to implement and provides a high-throughput solution. We predict that within the broad field of bioelectronic medicine, those teams that incorporate high-density neural recording devices to their armamentarium might find our framework quite valuable as they expand their analytical footprint.

17.
Nat Commun ; 11(1): 1403, 2020 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-32179753

RESUMEN

Patients with Systemic lupus erythematosus (SLE) experience various peripheral and central nervous system manifestations including spatial memory impairment. A subset of autoantibodies (DNRAbs) cross-react with the GluN2A and GluN2B subunits of the NMDA receptor (NMDAR). We find that these DNRAbs act as positive allosteric modulators on NMDARs with GluN2A-containing NMDARs, even those containing a single GluN2A subunit, exhibiting a much greater sensitivity to DNRAbs than those with exclusively GluN2B. Accordingly, GluN2A-specific antagonists provide greater protection from DNRAb-mediated neuronal cell death than GluN2B antagonists. Using transgenic mice to perturb expression of either GluN2A or GluN2B in vivo, we find that DNRAb-mediated disruption of spatial memory characterized by early neuronal cell death and subsequent microglia-dependent pathologies requires GluN2A-containing NMDARs. Our results indicate that GluN2A-specific antagonists or negative allosteric modulators are strong candidates to treat SLE patients with nervous system dysfunction.


Asunto(s)
Autoanticuerpos/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/psicología , Receptores de N-Metil-D-Aspartato/inmunología , Memoria Espacial , Animales , Muerte Celular , Femenino , Humanos , Lupus Eritematoso Sistémico/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/inmunología , Receptores de N-Metil-D-Aspartato/genética
18.
Sci Rep ; 10(1): 14446, 2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32879327

RESUMEN

The concept that exposure in utero to maternal anti-brain antibodies contributes to the development of autism spectrum disorders (ASD) has been entertained for over a decade. We determined that antibodies targeting Caspr2 are present at high frequency in mothers with brain-reactive serology and a child with ASD, and further demonstrated that exposure in utero to a monoclonal anti-Caspr2 antibody, derived from a mother of an ASD child, led to an-ASD like phenotype in male offspring. Now we propose a new model to study the effects of in utero exposure to anti-Caspr2 antibody. Dams immunized with the extracellular portion of Caspr2 express anti-Caspr2 antibodies throughout gestation to better mimic the human condition. Male but not female mice born to dams harboring polyclonal anti-Caspr2 antibodies showed abnormal cortical development, decreased dendritic complexity of excitatory neurons and reduced numbers of inhibitory neurons in the hippocampus, as well as repetitive behaviors and impairments in novelty interest in the social preference test as adults. These data supporting the pathogenicity of anti-Caspr2 antibodies are consistent with the concept that anti-brain antibodies present in women during gestation can alter fetal brain development, and confirm that males are peculiarly susceptible.


Asunto(s)
Trastorno del Espectro Autista/genética , Autoanticuerpos/inmunología , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Animales , Anticuerpos Antiidiotipos/genética , Anticuerpos Antiidiotipos/inmunología , Trastorno del Espectro Autista/inmunología , Trastorno del Espectro Autista/fisiopatología , Autoanticuerpos/efectos adversos , Conducta Animal , Encéfalo/inmunología , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Hipocampo/inmunología , Hipocampo/patología , Humanos , Masculino , Herencia Materna/genética , Herencia Materna/inmunología , Relaciones Materno-Fetales , Proteínas de la Membrana/inmunología , Ratones , Proteínas del Tejido Nervioso/inmunología , Neurogénesis/inmunología , Problema de Conducta
19.
J Neurosci Res ; 87(10): 2211-21, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19301430

RESUMEN

gamma-Aminobutyric acid (GABA) regulates the proliferation and migration of olfactory bulb (OB) interneuron progenitors derived from the subventricular zone (SVZ), but the role of GABA in the differentiation of these progenitors has been largely unexplored. This study examines the role of GABA in the differentiation of OB dopaminergic interneurons using neonatal forebrain organotypic slice cultures prepared from transgenic mice expressing green fluorescent protein (GFP) under the control of the tyrosine hydroxylase (Th) gene promoter (ThGFP). KCl-mediated depolarization of the slices induced ThGFP expression. The addition of GABA to the depolarized slices further increased GFP fluorescence by inducing ThGFP expression in an additional set of periglomerular cells. These findings show that GABA promoted differentiation of SVZ-derived OB dopaminergic interneurons and suggest that GABA indirectly regulated Th expression and OB dopaminergic neuron differentiation through an acceleration of the maturation rate for the dopaminergic progenitors. Additional studies revealed that the effect of GABA on ThGFP expression required activation of L- and P/Q-type Ca2+ channels as well as GABA(A) and GABA(B) receptors. These voltage-gated Ca2+ channels and GABA receptors have previously been shown to be required for the coexpressed GABAergic phenotype in the OB interneurons. Together, these findings suggest that Th expression and the differentiation of OB dopaminergic interneurons are coupled to the coexpressed GABAergic phenotype and demonstrate a novel role for GABA in neurogenesis.


Asunto(s)
Dopamina/metabolismo , Expresión Génica/efectos de los fármacos , Interneuronas/efectos de los fármacos , Bulbo Olfatorio/citología , Ácido gamma-Aminobutírico/farmacología , Agatoxinas , Aminoácidos/farmacología , Animales , Animales Recién Nacidos , Bicuculina/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas del GABA/farmacología , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Transgénicos , Ácidos Fosfínicos/farmacología , Cloruro de Potasio/farmacología , Prosencéfalo/citología , Prosencéfalo/metabolismo , Venenos de Araña/farmacología , Técnicas de Cultivo de Tejidos , Tirosina 3-Monooxigenasa/genética , Xantenos/farmacología
20.
J Neuroeng Rehabil ; 6: 7, 2009 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-19254380

RESUMEN

Transcranial magnetic stimulation (TMS) has quickly progressed from a technical curiosity to a bona-fide tool for neurological research. The impetus has been due to the promising results obtained when using TMS to uncover neural processes in normal human subjects, as well as in the treatment of intractable neurological conditions, such as stroke, chronic depression and epilepsy. The basic principle of TMS is that most neuronal axons that fall within the volume of magnetic stimulation become electrically excited, trigger action potentials and release neurotransmitter into the postsynaptic neurons. What happens afterwards remains elusive, especially in the case of repeated stimulation. Here we discuss the likelihood that certain TMS protocols produce long-term changes in cortical synapses akin to long-term potentiation and long-term depression of synaptic transmission. Beyond the synaptic effects, TMS might have consequences on other neuronal processes, such as genetic and protein regulation, and circuit-level patterns, such as network oscillations. Furthermore, TMS might have non-neuronal effects, such as changes in blood flow, which are still poorly understood.


Asunto(s)
Encéfalo/fisiología , Plasticidad Neuronal/fisiología , Sinapsis/fisiología , Estimulación Magnética Transcraneal , Humanos , Inhibición Neural/fisiología , Transmisión Sináptica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA