Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
EMBO J ; 40(5): e104267, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33491217

RESUMEN

Impairments in social relationships and awareness are features observed in autism spectrum disorders (ASDs). However, the underlying mechanisms remain poorly understood. Shank2 is a high-confidence ASD candidate gene and localizes primarily to postsynaptic densities (PSDs) of excitatory synapses in the central nervous system (CNS). We show here that loss of Shank2 in mice leads to a lack of social attachment and bonding behavior towards pubs independent of hormonal, cognitive, or sensitive deficits. Shank2-/- mice display functional changes in nuclei of the social attachment circuit that were most prominent in the medial preoptic area (MPOA) of the hypothalamus. Selective enhancement of MPOA activity by DREADD technology re-established social bonding behavior in Shank2-/- mice, providing evidence that the identified circuit might be crucial for explaining how social deficits in ASD can arise.


Asunto(s)
Trastorno Autístico/tratamiento farmacológico , Modelos Animales de Enfermedad , Relaciones Interpersonales , Conducta Materna/efectos de los fármacos , Proteínas del Tejido Nervioso/fisiología , Piperazinas/farmacología , Área Preóptica/efectos de los fármacos , Animales , Trastorno Autístico/etiología , Trastorno Autístico/metabolismo , Trastorno Autístico/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Área Preóptica/metabolismo , Área Preóptica/patología , Sinapsis
2.
Brain ; 146(12): 4916-4934, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37849234

RESUMEN

Alzheimer's disease is a complex neurodegenerative disorder leading to a decline in cognitive function and mental health. Recent research has positioned the gut microbiota as an important susceptibility factor in Alzheimer's disease by showing specific alterations in the gut microbiome composition of Alzheimer's patients and in rodent models. However, it is unknown whether gut microbiota alterations are causal in the manifestation of Alzheimer's symptoms. To understand the involvement of Alzheimer's patient gut microbiota in host physiology and behaviour, we transplanted faecal microbiota from Alzheimer's patients and age-matched healthy controls into microbiota-depleted young adult rats. We found impairments in behaviours reliant on adult hippocampal neurogenesis, an essential process for certain memory functions and mood, resulting from Alzheimer's patient transplants. Notably, the severity of impairments correlated with clinical cognitive scores in donor patients. Discrete changes in the rat caecal and hippocampal metabolome were also evident. As hippocampal neurogenesis cannot be measured in living humans but is modulated by the circulatory systemic environment, we assessed the impact of the Alzheimer's systemic environment on proxy neurogenesis readouts. Serum from Alzheimer's patients decreased neurogenesis in human cells in vitro and were associated with cognitive scores and key microbial genera. Our findings reveal for the first time, that Alzheimer's symptoms can be transferred to a healthy young organism via the gut microbiota, confirming a causal role of gut microbiota in Alzheimer's disease, and highlight hippocampal neurogenesis as a converging central cellular process regulating systemic circulatory and gut-mediated factors in Alzheimer's.


Asunto(s)
Enfermedad de Alzheimer , Microbioma Gastrointestinal , Humanos , Ratas , Animales , Hipocampo , Cognición , Microbioma Gastrointestinal/fisiología , Neurogénesis/fisiología
3.
Mol Psychiatry ; 27(12): 4994-5006, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36100669

RESUMEN

Members of the Shank protein family are master scaffolds of the postsynaptic architecture and mutations within the SHANK genes are causally associated with autism spectrum disorders (ASDs). We generated a Shank2-Shank3 double knockout mouse that is showing severe autism related core symptoms, as well as a broad spectrum of comorbidities. We exploited this animal model to identify cortical brain areas linked to specific autistic traits by locally deleting Shank2 and Shank3 simultaneously. Our screening of 10 cortical subregions revealed that a Shank2/3 deletion within the retrosplenial area severely impairs social memory, a core symptom of ASD. Notably, DREADD-mediated neuronal activation could rescue the social impairment triggered by Shank2/3 depletion. Data indicate that the retrosplenial area has to be added to the list of defined brain regions that contribute to the spectrum of behavioural alterations seen in ASDs.


Asunto(s)
Trastorno del Espectro Autista , Giro del Cíngulo , Interacción Social , Animales , Ratones , Trastorno del Espectro Autista/genética , Proteínas de Microfilamentos/genética , Mutación , Proteínas del Tejido Nervioso/genética , Neuronas/fisiología , Giro del Cíngulo/metabolismo , Giro del Cíngulo/patología
4.
Mol Psychiatry ; 26(8): 3778-3794, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32051550

RESUMEN

Older patients with severe physical trauma are at high risk of developing neuropsychiatric syndromes with global impairment of cognition, attention, and consciousness. We employed a thoracic trauma (TxT) mouse model and thoroughly analyzed age-dependent spatial and temporal posttraumatic alterations in the central nervous system. Up to 5 days after trauma, we observed a transient 50% decrease in the number of excitatory synapses specifically in hippocampal pyramidal neurons accompanied by alterations in attention and motor activity and disruption of contextual memory consolidation. In parallel, hippocampal corticotropin-releasing hormone (CRH) expression was highly upregulated, and brain-derived neurotrophic factor (BDNF) levels were significantly reduced. In vitro experiments revealed that CRH application induced neuronal autophagy with rapid lysosomal degradation of BDNF via the NF-κB pathway. The subsequent synaptic loss was rescued by BDNF as well as by specific NF-κB and CRH receptor 1 (CRHR1) antagonists. In vivo, the chronic application of a CRHR1 antagonist after TxT resulted in reversal of the observed histological, molecular, and behavioral alterations. The data suggest that neuropsychiatric syndromes (i.e., delirium) after peripheral trauma might be at least in part due to the activation of the hippocampal CRH/NF-κB/BDNF pathway, which results in a dramatic loss of synaptic contacts. The successful rescue by stress hormone receptor antagonists should encourage clinical trials focusing on trauma-induced delirium and/or other posttraumatic syndromes.


Asunto(s)
Delirio , Neuronas , Animales , Hormona Liberadora de Corticotropina , Humanos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Ratones , Receptores de Hormona Liberadora de Corticotropina , Síndrome
5.
Brain ; 137(Pt 1): 137-52, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24277719

RESUMEN

Proteins of the ProSAP/Shank family act as major organizing scaffolding elements within the postsynaptic density of excitatory synapses. Deletions, mutations or the downregulation of these molecules has been linked to autism spectrum disorders, the related Phelan McDermid Syndrome or Alzheimer's disease. ProSAP/Shank proteins are targeted to synapses depending on binding to zinc, which is a prerequisite for the assembly of the ProSAP/Shank scaffold. To gain insight into whether the previously reported assembly of ProSAP/Shank through zinc ions provides a crossing point between genetic forms of autism spectrum disorder and zinc deficiency as an environmental risk factor for autism spectrum disorder, we examined the interplay between zinc and ProSAP/Shank in vitro and in vivo using neurobiological approaches. Our data show that low postsynaptic zinc availability affects the activity dependent increase in ProSAP1/Shank2 and ProSAP2/Shank3 levels at the synapse in vitro and that a loss of synaptic ProSAP1/Shank2 and ProSAP2/Shank3 occurs in a mouse model for acute and prenatal zinc deficiency. Zinc-deficient animals displayed abnormalities in behaviour such as over-responsivity and hyperactivity-like behaviour (acute zinc deficiency) and autism spectrum disorder-related behaviour such as impairments in vocalization and social behaviour (prenatal zinc deficiency). Most importantly, a low zinc status seems to be associated with an increased incidence rate of seizures, hypotonia, and attention and hyperactivity issues in patients with Phelan-McDermid syndrome, which is caused by haploinsufficiency of ProSAP2/Shank3. We suggest that the molecular underpinning of prenatal zinc deficiency as a risk factor for autism spectrum disorder may unfold through the deregulation of zinc-binding ProSAP/Shank family members.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/metabolismo , Saposinas/metabolismo , Sinapsis/fisiología , Zinc/deficiencia , Animales , Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Conducta Animal/fisiología , Western Blotting , Células Cultivadas , Trastornos Generalizados del Desarrollo Infantil/fisiopatología , Deleción Cromosómica , Trastornos de los Cromosomas/metabolismo , Trastornos de los Cromosomas/fisiopatología , Cromosomas Humanos Par 22/metabolismo , Femenino , Hipocampo/metabolismo , Humanos , Inmunohistoquímica , Ratones , Técnicas de Cultivo de Órganos , Embarazo , ARN Interferente Pequeño/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Espectrometría de Fluorescencia , Transfección , Vocalización Animal/fisiología
6.
Mol Autism ; 14(1): 21, 2023 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-37316943

RESUMEN

BACKGROUND: Autism Spectrum Disorders (ASD) patients experience disturbed nociception in the form of either hyposensitivity to pain or allodynia. A substantial amount of processing of somatosensory and nociceptive stimulus takes place in the dorsal spinal cord. However, many of these circuits are not very well understood in the context of nociceptive processing in ASD. METHODS: We have used a Shank2-/- mouse model, which displays a set of phenotypes reminiscent of ASD, and performed behavioural and microscopic analysis to investigate the role of dorsal horn circuitry in nociceptive processing of ASD. RESULTS: We determined that Shank2-/- mice display increased sensitivity to formalin pain and thermal preference, but a sensory specific mechanical allodynia. We demonstrate that high levels of Shank2 expression identifies a subpopulation of neurons in murine and human dorsal spinal cord, composed mainly by glycinergic interneurons and that loss of Shank2 causes the decrease in NMDAR in excitatory synapses on these inhibitory interneurons. In fact, in the subacute phase of the formalin test, glycinergic interneurons are strongly activated in wild type (WT) mice but not in Shank2-/- mice. Consequently, nociception projection neurons in laminae I are activated in larger numbers in Shank2-/- mice. LIMITATIONS: Our investigation is limited to male mice, in agreement with the higher representation of ASD in males; therefore, caution should be applied to extrapolate the findings to females. Furthermore, ASD is characterized by extensive genetic diversity and therefore the findings related to Shank2 mutant mice may not necessarily apply to patients with different gene mutations. Since nociceptive phenotypes in ASD range between hyper- and hypo-sensitivity, diverse mutations may affect the circuit in opposite ways. CONCLUSION: Our findings prove that Shank2 expression identifies a new subset of inhibitory interneurons involved in reducing the transmission of nociceptive stimuli and whose unchecked activation is associated with pain hypersensitivity. We provide evidence that dysfunction in spinal cord pain processing may contribute to the nociceptive phenotypes in ASD.


Asunto(s)
Trastorno Autístico , Femenino , Humanos , Masculino , Animales , Ratones , Trastorno Autístico/genética , Nocicepción , Neuronas , Interneuronas , Dolor , Proteínas del Tejido Nervioso/genética
7.
J Med Food ; 25(9): 924-929, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34185600

RESUMEN

Middle age is increasingly accepted as a key period during which individuals are susceptible to the effect of environmental and lifestyle factors. Emerging research indicates that dietary factors play a crucial role in brain health and cognitive function, and studies in both animals and humans have demonstrated that dietary interventions can mitigate cognitive impairment. Specifically, magnesium has been shown to enhance learning and memory, and magnesium deficiency is associated with impaired hippocampal-dependent memory formation in animal studies. The aim of this study was to examine if supplementation with a magnesium-rich marine mineral blend (MMB) could alter middle-age-related cognitive impairment. Young and middle-aged rats were given access to a control diet or an experimental diet formulated with an MMB for 4 weeks before undergoing a series of behavioral assessments. Supplementation of MMB to middle-aged rats rescued a deficit in cognitive impairment, specifically a pattern separation paradigm that is sensitive to alterations in a type of brain plasticity called neurogenesis. It had no effect on general activity in the open field or performance on other hippocampal-associated tasks. Changes in cognitive function occur as a predictable consequence of aging. Research into whether modification of dietary factors, such as this MMB, may play a role in the prevention of age-related cognitive impairment warrants further investigation.


Asunto(s)
Hipocampo , Magnesio , Envejecimiento , Animales , Suplementos Dietéticos , Humanos , Trastornos de la Memoria , Persona de Mediana Edad , Minerales , Neurogénesis , Ratas
8.
Pharmaceutics ; 11(11)2019 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-31683907

RESUMEN

The accumulation of amyloid ß (Aß) triggers a cascade of toxic events in Alzheimer's disease (AD). The KLVFF peptide can interfere with Aß aggregation. However, the peptide suffers from poor bioavailability and the inability to cross the blood-brain barrier. In this work, we study the possibility of adopting nanomedicine to overcome KLVFF limits in biodistribution. We produced new engineered polymeric nanoparticles (NPs), and we evaluated the cellular toxicity of these NPs and validated that KVLFF peptides released by NPs show the same promising effects on AD pathology. Our results revealed the successful generation of KVLFF loaded NPs that, without significant effects on cell heath, are even more potent in reversing Aß-induced pathologies compared to the free peptide. Therefore, NPs will significantly advance KVLFF treatment as a therapeutic option for AD.

9.
Front Mol Neurosci ; 10: 180, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28642683

RESUMEN

RICH2 knockout (RICH2 KO) mice exhibit neophobia in the novel object test. To gain further insight into their anxiety-related phenotype, we subjected these mice to additional behavioral tests to elucidate whether the behavioral abnormality in these mice is a consequence of reduced exploratory motivation, and whether the neophobia is linked specifically to objects or also present for other modalities. RICH2 KO mice engage in normal exploration in a novel environment, suggesting that the anxiety-related phenotype is not due to reduced exploratory drive. Increased fear response was not observed using novel olfactory cues, but restricted to objects. Given that the amygdala is an important brain region mediating anxiety-related behaviors and a prime target for anxiety-related therapeutics, and RICH2 is a Rho-GTPase activating protein (GAP) regulating synaptic spine plasticity via small GTPases, we analyzed spine formation, morphology and receptor composition in amygdala. We found disinhibition of RhoA in the amygdala of RICH2 KO mice, along with a decreased ability for actin polymerization and a reduction in mature spines. However, we detected increased neuronal activation in the amygdala evidenced by c-fos labeling. Thus, we conclude that despite unaltered baseline activity, RICH2 KO mice show heightened amygdala response after exposure to objects, which, however, does not result in homeostatic strengthening of excitatory synapses.

10.
Front Mol Neurosci ; 10: 450, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29379414

RESUMEN

A number of studies have reported changes in the hemispheric dominance in autism spectrum disorder (ASD) patients on functional, biochemical, and morphological level. Since asymmetry of the brain is also found in many vertebrates, we analyzed whether prenatal zinc deficient (PZD) mice, a mouse model with ASD like behavior, show alterations regarding brain lateralization on molecular and behavioral level. Our results show that hemisphere-specific expression of marker genes is abolished in PZD mice on mRNA and protein level. Using magnetic resonance imaging, we found an increased striatal volume in PZD mice with no change in total brain volume. Moreover, behavioral patterns associated with striatal lateralization are altered and the lateralized expression of dopamine receptor 1 (DR1) in the striatum of PZD mice was changed. We conclude that zinc signaling during brain development has a critical role in the establishment of brain lateralization in mice.

11.
Front Behav Neurosci ; 10: 37, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26973485

RESUMEN

Zinc deficiency has recently been linked to the etiology of autism spectrum disorders (ASD) as environmental risk factor. With an estimated 17% of the world population being at risk of zinc deficiency, especially zinc deficiency during pregnancy might be a common occurrence, also in industrialized nations. On molecular level, zinc deficiency has been shown to affect a signaling pathway at glutamatergic synapses that has previously been identified through genetic mutations in ASD patients, the Neurexin-Neuroligin-Shank pathway, via altering zinc binding Shank family members. In particular, prenatal zinc deficient but not acute zinc deficient animals have been reported to display autism like behavior in some behavioral tests. However, a full behavioral analysis of a possible autism like behavior has been lacking so far. Here, we performed an extensive behavioral phenotyping of mice born from mothers with mild zinc deficiency during all trimesters of pregnancy. Prenatal zinc deficient animals were investigated as adults and gender differences were assessed. Our results show that prenatal zinc deficient mice display increased anxiety, deficits in nest building and various social interaction paradigm, as well as mild alterations in ultrasonic vocalizations. A gender specific analysis revealed only few sex specific differences. Taken together, given that similar behavioral abnormalities as reported here are frequently observed in ASD mouse models, we conclude that prenatal zinc deficient animals even without specific genetic susceptibility for ASD, already show some features of ASD like behavior.

12.
Mol Brain ; 9: 28, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26969129

RESUMEN

BACKGROUND: The majority of neurons within the central nervous system receive their excitatory inputs via small, actin-rich protrusions called dendritic spines. Spines can undergo rapid morphological alterations according to synaptic activity. This mechanism is implicated in learning and memory formation as it is ultimately altering the number and distribution of receptors and proteins at the post-synaptic membrane, thereby regulating synaptic input. The Rho-family GTPases play an important role in regulating this spine plasticity by the interaction with cytoskeletal components and several signaling pathways within the spine compartment. Rho-GAP interacting CIP4 homologue2/RICH2 is a Rho-GAP protein regulating small GTPases and was identified as an interaction partner of the scaffolding protein SHANK3 at post-synaptic densities. RESULTS: Here, we characterize the loss of RICH2 in a novel mouse model. Our results show that RICH2 KO animals display a selective and highly significant fear of novel objects and increased stereotypic behavior as well as impairment of motor learning. We found an increase in multiple spine synapses in the hippocampus and cerebellum along with alterations in receptor composition and actin polymerization. Furthermore, we observed that the loss of RICH2 leads to a disinhibition of synaptic RAC1 in vivo. CONCLUSIONS: The results are in line with the reported role of RAC1 activity being essential for activity-dependent spine enlargement. Since SHANK3 mutations are known to be causative for neuropsychiatric diseases of the Autism Spectrum (ASD), a disintegrated SHANK3/RICH2 complex at synaptic sites might at least in part be responsible for abnormal spine formation and plasticity in ASDs.


Asunto(s)
Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Proteínas Activadoras de GTPasa/deficiencia , Trastornos Fóbicos/metabolismo , Animales , Conducta Animal , Glutamatos/metabolismo , Ratones Mutantes , Actividad Motora , Trastornos Fóbicos/fisiopatología , Transducción de Señal , Sinapsis/metabolismo
13.
Exp Neurol ; 253: 126-37, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24382453

RESUMEN

Recently, mutations in ProSAP2/Shank3 have been discovered as one of the genetic factors for schizophrenia (SCZ). Here, we show that the postsynaptic density protein ProSAP2/Shank3 undergoes activity dependent synapse-to-nucleus shuttling in hippocampal neurons. Our study shows that the de novo mutation (R1117X) in ProSAP2/Shank3 that was identified in a patient with SCZ leads to an accumulation of mutated ProSAP2/Shank3 within the nucleus independent of synaptic activity. Furthermore, we identified novel nuclear ProSAP2/Shank3 interaction partners. Nuclear localization of mutated ProSAP2/Shank3 alters transcription of several genes, among them already identified genetic risk factors for SCZ such as Synaptotagmin 1 and LRRTM1. Comparing the SCZ mutation of ProSAP2/Shank3 to the knockdown of ProSAP2/Shank3 we found some shared features such as reduced synaptic density in neuronal cultures. However, hippocampal neurons expressing the ProSAP2/Shank3 SCZ mutation furthermore show altered E/I ratio and reduced dendritic branching. Thus, we conclude that the uncoupling of ProSAP2/Shank3 nuclear shuttling from synaptic activity may represent a molecular mechanism that contributes to the pathology of SCZ in patients with mutations in ProSAP2/Shank3.


Asunto(s)
Núcleo Celular/metabolismo , Regulación de la Expresión Génica/genética , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo , Animales , Arginina/genética , Células COS , Células Cultivadas , Chlorocebus aethiops , Embrión de Mamíferos , Hipocampo/citología , Humanos , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neuronas/ultraestructura , Ratas , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA