Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
1.
Brain Behav Immun ; 117: 493-509, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38307446

RESUMEN

In the last years, the hypothesis that elevated levels of proinflammatory cytokines contribute to the pathogenesis of neurodevelopmental diseases has gained popularity. IL-1 is one of the main cytokines found to be elevated in Autism spectrum disorder (ASD), a complex neurodevelopmental condition characterized by defects in social communication and cognitive impairments. In this study, we demonstrate that mice lacking IL-1 signaling display autistic-like defects associated with an excessive number of synapses. We also show that microglia lacking IL-1 signaling at early neurodevelopmental stages are unable to properly perform the process of synapse engulfment and display excessive activation of mammalian target of rapamycin (mTOR) signaling. Notably, even the acute inhibition of IL-1R1 by IL-1Ra is sufficient to enhance mTOR signaling and reduce synaptosome phagocytosis in WT microglia. Finally, we demonstrate that rapamycin treatment rescues the defects in IL-1R deficient mice. These data unveil an exclusive role of microglial IL-1 in synapse refinement via mTOR signaling and indicate a novel mechanism possibly involved in neurodevelopmental disorders associated with defects in the IL-1 pathway.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Animales , Ratones , Microglía , Serina-Treonina Quinasas TOR , Citocinas , Sirolimus/farmacología , Sinapsis , Interleucina-1 , Mamíferos
2.
Neurobiol Dis ; 192: 106416, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38272141

RESUMEN

BACKGROUND: The dysregulation of the gut-brain axis in chronic inflammatory bowel diseases can cause neuro-psychological disturbances, but the underlying mechanisms are still not fully understood. The choroid plexus (CP) maintains brain homeostasis and nourishment through the secretion and clearance of cerebrospinal fluid. Recent research has demonstrated the existence of a CP vascular barrier in mice which is modulated during intestinal inflammation. This study investigates possible correlations between CP modifications and inflammatory activity in patients with Crohn's disease (CD). METHODS: In this prospective study, 17 patients with CD underwent concomitant abdominal and brain 3 T MRI. The volume and permeability of CP were compared with levels of C-reactive protein (CRP), fecal calprotectin (FC), sMARIA and SES-CD scores. RESULTS: The CP volume was negatively correlated with CRP levels (R = -0.643, p-value = 0.024) and FC (R = -0.571, p-value = 0.050). DCE metrics normalized by CP volume were positively correlated with CRP (K-trans: R = 0.587, p-value = 0.045; Vp: R = 0.706, p-value = 0.010; T1: R = 0.699, p-value = 0.011), and FC (Vp: R = 0.606, p-value = 0.037). CONCLUSIONS: Inflammatory activity in patients with CD is associated with changes in CP volume and permeability, thus supporting the hypothesis that intestinal inflammation could affect the brain through the modulation of CP vascular barrier also in humans.


Asunto(s)
Enfermedad de Crohn , Humanos , Animales , Ratones , Enfermedad de Crohn/diagnóstico por imagen , Enfermedad de Crohn/metabolismo , Plexo Coroideo/diagnóstico por imagen , Plexo Coroideo/metabolismo , Estudios Prospectivos , Eje Cerebro-Intestino , Biomarcadores/metabolismo , Proteína C-Reactiva/análisis , Proteína C-Reactiva/metabolismo , Complejo de Antígeno L1 de Leucocito/metabolismo , Índice de Severidad de la Enfermedad , Inflamación/diagnóstico por imagen , Permeabilidad
3.
Immunity ; 57(1): 86-105.e9, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38159572

RESUMEN

Triggering receptor expressed on myeloid cells 2 (Trem2) is a myeloid cell-specific gene expressed in brain microglia, with variants that are associated with neurodegenerative diseases, including Alzheimer's disease. Trem2 is essential for microglia-mediated synaptic refinement, but whether Trem2 contributes to shaping neuronal development remains unclear. Here, we demonstrate that Trem2 plays a key role in controlling the bioenergetic profile of pyramidal neurons during development. In the absence of Trem2, developing neurons in the hippocampal cornus ammonis (CA)1 but not in CA3 subfield displayed compromised energetic metabolism, accompanied by reduced mitochondrial mass and abnormal organelle ultrastructure. This was paralleled by the transcriptional rearrangement of hippocampal pyramidal neurons at birth, with a pervasive alteration of metabolic, oxidative phosphorylation, and mitochondrial gene signatures, accompanied by a delay in the maturation of CA1 neurons. Our results unveil a role of Trem2 in controlling neuronal development by regulating the metabolic fitness of neurons in a region-specific manner.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Metabolismo Energético , Microglía/metabolismo , Neuronas/metabolismo , Animales , Ratones
4.
Biochem Pharmacol ; 213: 115633, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37269887

RESUMEN

Neratinib (NE) is an irreversible pan-ERBB tyrosine kinase inhibitor used to treat breast cancers (BCa) with amplification of the ERBB2/HER2/Neu gene or overexpression of the ERBB2 receptor. However, the mechanisms behind this process are not fully understood. Here we investigated the effects of NE on critical cell survival processes in ERBB2+ cancer cells. By kinome array analysis, we showed that NE time-dependently inhibited the phosphorylation of two distinct sets of kinases. The first set, including ERBB2 downstream signaling kinases such as ERK1/2, ATK, and AKT substrates, showed inhibition after 2 h of NE treatment. The second set, which comprised kinases involved in DNA damage response, displayed inhibition after 72 h. Flow cytometry analyses showed that NE induced G0/G1 cell cycle arrest and early apoptosis. By immunoblot, light and electron microscopy, we revealed that NE also transiently induced autophagy, mediated by increased expression levels and nuclear localization of TFEB and TFE3. Altered TFEB/TFE3 expression was accompanied by dysregulation of mitochondrial energy metabolism and dynamics, leading to a decrease in ATP production, glycolytic activity, and a transient downregulation of fission proteins. Increased TFEB and TFE3 expression was also observed in ERBB2-/ERBB1 + BCa cells, supporting that NE may act through other ERBB family members and/or other kinases. Overall, this study highlights NE as a potent activator of TFEB and TFE3, leading to the suppression of cancer cell survival through autophagy induction, cell cycle arrest, apoptosis, mitochondrial dysfunction and inhibition of DNA damage response.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Autofagia , Metabolismo Energético
5.
EMBO J ; 42(13): e113796, 2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37161785

RESUMEN

In the last two decades, the term synaptopathy has been largely used to underline the concept that impairments of synaptic structure and function are the major determinant of brain disorders, including neurodevelopmental disorders. This notion emerged from the progress made in understanding the genetic architecture of neurodevelopmental disorders, which highlighted the convergence of genetic risk factors onto molecular pathways specifically localized at the synapse. However, the multifactorial origin of these disorders also indicated the key contribution of environmental factors. It is well recognized that inflammation is a risk factor for neurodevelopmental disorders, and several immune molecules critically contribute to synaptic dysfunction. In the present review, we highlight this concept, which we define by the term "immune-synaptopathy," and we discuss recent evidence suggesting a bi-directional link between the genetic architecture of individuals and maternal activation of the immune system in modulating brain developmental trajectories in health and disease.


Asunto(s)
Trastornos del Neurodesarrollo , Humanos , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/metabolismo , Sinapsis/metabolismo , Familia
7.
Ageing Res Rev ; 86: 101867, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36720351

RESUMEN

The study of pollutant effects is extremely important to address the epochal challenges we are facing, where world populations are increasingly moving from rural to urban centers, revolutionizing our world into an urban world. These transformations will exacerbate pollution, thus highlighting the necessity to unravel its effect on human health. Epidemiological studies have reported that pollution increases the risk of neurological diseases, with growing evidence on the risk of neurodegenerative disorders. Air pollution and water pollutants are the main chemicals driving this risk. These chemicals can promote inflammation, acting in synergy with genotype vulnerability. However, the biological underpinnings of this association are unknown. In this review, we focus on the link between pollution and brain network connectivity at the macro-scale level. We provide an updated overview of epidemiological findings and studies investigating brain network changes associated with pollution exposure, and discuss the mechanistic insights of pollution-induced brain changes through neural networks. We explain, in detail, the pollutome-connectome axis that might provide the functional substrate for pollution-induced processes leading to cognitive impairment and neurodegeneration. We describe this model within the framework of two pollutants, air pollution, a widely recognized threat, and polyfluoroalkyl substances, a large class of synthetic chemicals which are currently emerging as new neurotoxic source.


Asunto(s)
Contaminación del Aire , Disfunción Cognitiva , Conectoma , Enfermedades Neurodegenerativas , Humanos , Contaminación del Aire/efectos adversos , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/epidemiología , Inflamación
8.
Commun Biol ; 5(1): 1276, 2022 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-36414721

RESUMEN

We examined effects of exposing female and male mice for 33 weeks to 45% or 60% high fat diet (HFD). Males fed with either diet were more vulnerable than females, displaying higher and faster increase in body weight and more elevated cholesterol and liver enzymes levels. Higher glucose metabolism was revealed by PET in the olfactory bulbs of both sexes. However, males also displayed altered anterior cortex and cerebellum metabolism, accompanied by a more prominent brain inflammation relative to females. Although both sexes displayed reduced transcripts of neuronal and synaptic genes in anterior cortex, only males had decreased protein levels of AMPA and NMDA receptors. Oppositely, to anterior cortex, cerebellum of HFD-exposed mice displayed hypometabolism and transcriptional up-regulation of neuronal and synaptic genes. These results indicate that male brain is more susceptible to metabolic changes induced by HFD and that the anterior cortex versus cerebellum display inverse susceptibility to HFD.


Asunto(s)
Dieta Alta en Grasa , Obesidad , Animales , Ratones , Masculino , Femenino , Dieta Alta en Grasa/efectos adversos , Obesidad/metabolismo , Encéfalo/metabolismo , Peso Corporal , Neuronas/metabolismo
9.
EMBO J ; 41(23): e111192, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36314682

RESUMEN

Intracerebral hemorrhages are recognized risk factors for neurodevelopmental disorders and represent early biomarkers for cognitive dysfunction and mental disability, but the pathways leading to their occurrence are not well defined. We report that a single intrauterine exposure of the immunostimulant Poly I:C to pregnant mice at gestational day 9, which models a prenatal viral infection and the consequent maternal immune activation, induces the defective formation of brain vessels and causes intracerebral hemorrhagic events, specifically in male offspring. We demonstrate that maternal immune activation promotes the production of the TGF-ß1 active form and the consequent enhancement of pSMAD1-5 in males' brain endothelial cells. TGF-ß1, in combination with IL-1ß, reduces the endothelial expression of CD146 and claudin-5, alters the endothelium-pericyte interplay resulting in low pericyte coverage, and increases hemorrhagic events in the adult offspring. By showing that exposure to Poly I:C at the beginning of fetal cerebral angiogenesis results in sex-specific alterations of brain vessels, we provide a mechanistic framework for the association between intragravidic infections and anomalies of the neural vasculature, which may contribute to neuropsychiatric disorders.


Asunto(s)
Hemorragia Cerebral , Efectos Tardíos de la Exposición Prenatal , Animales , Femenino , Masculino , Ratones , Embarazo , Conducta Animal , Encéfalo/irrigación sanguínea , Encéfalo/patología , Hemorragia Cerebral/patología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Poli I-C/efectos adversos , Efectos Tardíos de la Exposición Prenatal/patología , Factor de Crecimiento Transformador beta1/metabolismo
10.
Methods Mol Biol ; 2492: 225-240, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35733047

RESUMEN

An in vitro blood-brain barrier (BBB) model must be highly reproducible and imitate as much as possible the properties of the in vivo environment, from both the functional and anatomical point of view. In our latest work, a BBB prototype was implemented through the use of human primary brain cells and then integrated in a microfluidic platform (Lauranzano et al., Adv Biosyst 3:e1800335, 2019). Here we describe, step by step, the setting of a customized bio-mimetic platform, which uses human brain endothelial cells and primary astrocytic cells to allow the study of the complex interactions between the immune system and the brain in healthy and neuroinflammatory conditions. The model can be exploited to investigate the neuroimmune communication at the blood-brain interface and to examine the transmigration of patient-derived lymphocytes in order to envisage cutting-edge strategies to restore barrier integrity and block the immune cell influx into the CNS.


Asunto(s)
Astrocitos , Barrera Hematoencefálica , Transporte Biológico , Células Endoteliales , Humanos , Microfluídica
11.
FEBS J ; 289(12): 3374-3392, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-33998773

RESUMEN

Intragravidic and perinatal infections, acting through either direct viral effect or immune-mediated responses, are recognized causes of liability for neurodevelopmental disorders in the progeny. The large amounts of epidemiological data and the wealth of information deriving from animal models of gestational infections have contributed to delineate, in the last years, possible underpinning mechanisms for this phenomenon, including defects in neuronal migration, impaired spine and synaptic development, and altered activation of microglia. Recently, dysfunctions of the neurovascular unit and anomalies of the brain vasculature have unexpectedly emerged as potential causes at the origin of behavioral abnormalities and psychiatric disorders consequent to prenatal and perinatal infections. This review aims to discuss the up-to-date literature evidence pointing to the neurovascular unit and brain vasculature damages as the etiological mechanisms in neurodevelopmental syndromes. We focus on the inflammatory events consequent to intragravidic viral infections as well as on the direct viral effects as the potential primary triggers. These authors hope that a timely review of the literature will help to envision promising research directions, also relevant for the present and future COVID-19 longitudinal studies.


Asunto(s)
COVID-19 , Trastornos Mentales , Complicaciones Infecciosas del Embarazo , Efectos Tardíos de la Exposición Prenatal , Animales , Encéfalo , Femenino , Humanos , Embarazo
12.
Immunity ; 54(11): 2611-2631.e8, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34758338

RESUMEN

Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.


Asunto(s)
Hipocampo/metabolismo , Interleucina-6/biosíntesis , Exposición Materna , Neuronas/metabolismo , Efectos Tardíos de la Exposición Prenatal , Sinapsis/metabolismo , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Hipocampo/fisiopatología , Mediadores de Inflamación/metabolismo , Ratones , Embarazo , Transducción de Señal , Transmisión Sináptica
13.
Science ; 374(6566): 439-448, 2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34672740

RESUMEN

Up to 40% of patients with inflammatory bowel disease present with psychosocial disturbances. We previously identified a gut vascular barrier that controls the dissemination of bacteria from the intestine to the liver. Here, we describe a vascular barrier in the brain choroid plexus (PVB) that is modulated in response to intestinal inflammation through bacteria-derived lipopolysaccharide. The inflammatory response induces PVB closure after gut vascular barrier opening by the up-regulation of the wingless-type, catenin-beta 1 (Wnt/ß-catenin) signaling pathway, rendering it inaccessible to large molecules. In a model of genetically driven closure of choroid plexus endothelial cells, we observed a deficit in short-term memory and anxiety-like behavior, suggesting that PVB closure may correlate with mental deficits. Inflammatory bowel disease­related mental symptoms may thus be the consequence of a deregulated gut­brain vascular axis.


Asunto(s)
Plexo Coroideo/irrigación sanguínea , Plexo Coroideo/fisiopatología , Colitis Ulcerosa/fisiopatología , Colitis Ulcerosa/psicología , Intestinos/fisiopatología , Trastornos de la Memoria/fisiopatología , Memoria a Corto Plazo , Animales , Ansiedad/etiología , Ansiedad/fisiopatología , Barrera Hematoencefálica/patología , Colitis Ulcerosa/complicaciones , Dextranos , Modelos Animales de Enfermedad , Humanos , Lipopolisacáridos , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Microglía/patología , Transducción de Señal , Uniones Estrechas/patología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
14.
Neurooncol Adv ; 3(1): vdab076, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34377986

RESUMEN

BACKGROUND: The radio- and chemo-resistance of glioblastoma stem-like cells (GSCs), together with their innate tumor-initiating aptitude, make this cell population a crucial target for effective therapies. However, targeting GSCs is hardly difficult and complex, due to the presence of the blood-brain barrier (BBB) and the infiltrative nature of GSCs arousing their dispersion within the brain parenchyma. METHODS: Liposomes (LIPs), surface-decorated with an Apolipoprotein E-modified peptide (mApoE) to enable BBB crossing, were loaded with doxorubicin (DOXO), as paradigm of cytotoxic drug triggering immunogenic cell death (ICD). Patient-derived xenografts (PDXs) obtained by GSC intracranial injection were treated with mApoE-DOXO-LIPs alone or concomitantly with radiation. RESULTS: Our results indicated that mApoE, through the engagement of the low-density lipoprotein receptor (LDLR), promotes mApoE-DOXO-LIPs transcytosis across the BBB and confers target specificity towards GSCs. Irradiation enhanced LDLR expression on both BBB and GSCs, thus further promoting LIP diffusion and specificity. When administered in combination with radiations, mApoE-DOXO-LIPs caused a significant reduction of in vivo tumor growth due to GSC apoptosis. GSC apoptosis prompted microglia/macrophage phagocytic activity, together with the activation of the antigen-presenting machinery crucially required for anti-tumor adaptive immune response. CONCLUSIONS: Our results advocate for radiotherapy and adjuvant administration of drug-loaded, mApoE-targeted nanovectors as an effective strategy to deliver cytotoxic molecules to GSCs at the surgical tumor margins, the forefront of glioblastoma (GBM) recurrence, circumventing BBB hurdles. DOXO encapsulation proved in situ immune response activation within GBM microenvironment.

15.
Brain Behav Immun ; 98: 234-244, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34418501

RESUMEN

A deterioration in cognitive performance accompanies brain aging, even in the absence of neurodegenerative pathologies. However, the rate of cognitive decline can be slowed down by enhanced cognitive and sensorimotor stimulation protocols, such as environmental enrichment (EE). Understanding how EE exerts its beneficial effects on the aged brain pathophysiology can help in identifying new therapeutic targets. In this regard, the inflammatory chemokine ccl11/eotaxin-1 is a marker of aging with a strong relevance for neurodegenerative processes. Here, we demonstrate that EE in both elderly humans and aged mice decreases circulating levels of ccl11. Interfering, in mice, with the ccl11 decrease induced by EE ablated the beneficial effects on long-term memory retention, hippocampal neurogenesis, activation of local microglia and of ribosomal protein S6. On the other hand, treatment of standard-reared aged mice with an anti-ccl11 antibody resulted in EE-like improvements in spatial memory, hippocampal neurogenesis, and microglial activation. Taken together, our findings point to a decrease in circulating ccl11 concentration as a key mediator of the enhanced hippocampal function resulting from exposure to EE.


Asunto(s)
Ambiente , Hipocampo , Animales , Quimiocina CCL11 , Ratones , Neurogénesis , Memoria Espacial
16.
Cancers (Basel) ; 13(12)2021 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-34205341

RESUMEN

Therapeutic resistance after multimodal therapy is the most relevant cause of glioblastoma (GBM) recurrence. Extensive cellular heterogeneity, mainly driven by the presence of GBM stem-like cells (GSCs), strongly correlates with patients' prognosis and limited response to therapies. Defining the mechanisms that drive stemness and control responsiveness to therapy in a GSC-specific manner is therefore essential. Here we investigated the role of integrin a6 (ITGA6) in controlling stemness and resistance to radiotherapy in proneural and mesenchymal GSCs subtypes. Using cell sorting, gene silencing, RNA-Seq, and in vitro assays, we verified that ITGA6 expression seems crucial for proliferation and stemness of proneural GSCs, while it appears not to be relevant in mesenchymal GSCs under basal conditions. However, when challenged with a fractionated protocol of radiation therapy, comparable to that used in the clinical setting, mesenchymal GSCs were dependent on integrin a6 for survival. Specifically, GSCs with reduced levels of ITGA6 displayed a clear reduction of DNA damage response and perturbation of cell cycle pathways. These data indicate that ITGA6 inhibition is able to overcome the radioresistance of mesenchymal GSCs, while it reduces proliferation and stemness in proneural GSCs. Therefore, integrin a6 controls crucial characteristics across GBM subtypes in GBM heterogeneous biology and thus may represent a promising target to improve patient outcomes.

17.
Front Immunol ; 12: 640937, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33708226

RESUMEN

The role of microglia in controlling synapse homeostasis is becoming increasingly recognized by the scientific community. In particular, the microglia-mediated elimination of supernumerary synapses during development lays the basis for the correct formation of neuronal circuits in adulthood, while the possible reactivation of this process in pathological conditions, such as schizophrenia or Alzheimer's Disease, provides a promising target for future therapeutic strategies. The methodological approaches to investigate microglial synaptic engulfment include different in vitro and in vivo settings. Basic in vitro assays, employing isolated microglia and microbeads, apoptotic membranes, liposomes or synaptosomes allow the quantification of the microglia phagocytic abilities, while co-cultures of microglia and neurons, deriving from either WT or genetically modified mice models, provide a relatively manageable setting to investigate the involvement of specific molecular pathways. Further detailed analysis in mice brain is then mandatory to validate the in vitro assays as representative for the in vivo situation. The present review aims to dissect the main technical approaches to investigate microglia-mediated phagocytosis of neuronal and synaptic substrates in critical developmental time windows.


Asunto(s)
Microglía , Plasticidad Neuronal , Neurociencias/métodos , Sinapsis , Animales , Humanos , Neurogénesis/fisiología , Fagocitosis/fisiología
18.
EMBO Mol Med ; 13(3): e13785, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33555120

RESUMEN

The bidirectional link between heart and brain has intrigued scientists for ages, but little is known on the underlying mechanism. In their recent study, Fischer and colleagues (Islam et al, 2021) propose a mechanism by which heart failure-induced cognitive decline is linked to epigenetic changes that affect gene expression in neurons of hippocampus.


Asunto(s)
Insuficiencia Cardíaca , Hipocampo , Epigénesis Genética , Epigenómica , Insuficiencia Cardíaca/genética , Humanos , Neuronas
19.
Immunity ; 54(2): 191-193, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33567256

RESUMEN

The immune system actively regulates brain activity through the engagement of immune cells and immunomodulatory molecules. In this issue of Immunity, Klawonn et al. show that the activation of microglia in the striatum triggers an IL-6-mediated autocrine loop and the release of prostaglandins, which in turn induce a negative affective state via the stimulation of medium spiny neurons.


Asunto(s)
Microglía , Neuronas
20.
JCI Insight ; 6(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33373327

RESUMEN

Impairment of the GABAergic system has been reported in epilepsy, autism, attention deficit hyperactivity disorder, and schizophrenia. We recently demonstrated that ataxia telangiectasia mutated (ATM) directly shapes the development of the GABAergic system. Here, we show for the first time to our knowledge how the abnormal expression of ATM affects the pathological condition of autism. We exploited 2 different animal models of autism, the methyl CpG binding protein 2-null (Mecp2y/-) mouse model of Rett syndrome and mice prenatally exposed to valproic acid, and found increased ATM levels. Accordingly, treatment with the specific ATM kinase inhibitor KU55933 (KU) normalized molecular, functional, and behavioral defects in these mouse models, such as (a) delayed GABAergic development, (b) hippocampal hyperexcitability, (c) low cognitive performances, and (d) social impairments. Mechanistically, we demonstrate that KU administration to WT hippocampal neurons leads to (a) higher early growth response 4 activity on Kcc2b promoter, (b) increased expression of Mecp2, and (c) potentiated GABA transmission. These results provide evidence and molecular substrates for the pharmacological development of ATM inhibition in autism spectrum disorders.


Asunto(s)
Trastorno del Espectro Autista/tratamiento farmacológico , Animales , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Trastorno del Espectro Autista/fisiopatología , Trastorno del Espectro Autista/psicología , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Reparación del ADN , Modelos Animales de Enfermedad , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Masculino , Proteína 2 de Unión a Metil-CpG/deficiencia , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfolinas/farmacología , Embarazo , Efectos Tardíos de la Exposición Prenatal , Inhibidores de Proteínas Quinasas/farmacología , Pironas/farmacología , Síndrome de Rett/tratamiento farmacológico , Síndrome de Rett/fisiopatología , Síndrome de Rett/psicología , Simportadores/genética , Simportadores/metabolismo , Ácido Valproico/toxicidad , Cotransportadores de K Cl
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...