Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Neurochem ; 148(2): 168-187, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30144068

RESUMO

S100B is a Ca2+ -binding protein mainly concentrated in astrocytes. Its levels in biological fluids (cerebrospinal fluid, peripheral and cord blood, urine, saliva, amniotic fluid) are recognized as a reliable biomarker of active neural distress. Although the wide spectrum of diseases in which the protein is involved (acute brain injury, neurodegenerative diseases, congenital/perinatal disorders, psychiatric disorders) reduces its specificity, its levels remain an important aid in monitoring the trend of the disorder. Mounting evidence now points to S100B as a Damage-Associated Molecular Pattern molecule which, when released at high concentration, through its Receptor for Advanced Glycation Endproducts, triggers tissue reaction to damage in a series of different neural disorders. This review addresses this novel scenario, presenting data indicating that S100B levels and/or distribution in the nervous tissue of patients and/or experimental models of different neural disorders, for which the protein is used as a biomarker, are directly related to the progress of the disease: acute brain injury (ischemic/hemorrhagic stroke, traumatic injury), neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis), congenital/perinatal disorders (Down syndrome, spinocerebellar ataxia-1), psychiatric disorders (schizophrenia, mood disorders), inflammatory bowel disease. In many cases, over-expression/administration of the protein induces worsening of the disease, whereas its deletion/inactivation produces amelioration. This review points out that the pivotal role of the protein resulting from these data, opens the perspective that S100B may be regarded as a therapeutic target for these different diseases, which appear to share some common features reasonably attributable to neuroinflammation, regardless their origin.


Assuntos
Biomarcadores , Doenças do Sistema Nervoso , Subunidade beta da Proteína Ligante de Cálcio S100 , Animais , Humanos
2.
Neurochem Res ; 41(7): 1559-69, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26915108

RESUMO

Reelin is an extracellular matrix glycoprotein involved in the modulation of synaptic plasticity and essential for the proper radial migration of cortical neurons during development and for the integration and positioning of dentate granular cell progenitors; its expression is down-regulated as brain maturation is completed. Trimethyltin (TMT) is a potent neurotoxicant which causes selective neuronal death mainly localised in the CA1-CA3/hilus hippocampal regions. In the present study we analysed the expression of reelin and the modulation of endogenous neurogenesis in the postnatal rat hippocampus during TMT-induced neurodegeneration (TMT 6 mg/kg). Our results show that TMT administration induces changes in the physiological postnatal decrease of reelin expression in the hippocampus of developing rats. In particular, quantitative analysis of reelin-positive cells evidenced, in TMT-treated animals, a persistent reelin expression in the stratum lacunosum moleculare of Cornu Ammonis and in the molecular layer of Dentate Gyrus. In addition, a significant decrease in the number of bromodeoxyuridine (BrdU)-labeled newly-generated cells was also detectable in the subgranular zone of P21 TMT-treated rats compared with P21 control animals; no differences between P28 TMT-treated rats and age-matched control group were observed. In addition the neuronal commitment of BrdU-positive cells appeared reduced in P21 TMT-treated rats compared with P28 TMT-treated animals. Thus TMT treatment, administrated during development, induces an early reduction of endogenous neurogenesis and influences the hippocampal pattern of reelin expression in a temporally and regionally specific manner, altering the physiological decrease of this protein.


Assuntos
Moléculas de Adesão Celular Neuronais/biossíntese , Proteínas da Matriz Extracelular/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Neurogênese/fisiologia , Serina Endopeptidases/biossíntese , Compostos de Trimetilestanho/farmacologia , Animais , Animais Recém-Nascidos , Moléculas de Adesão Celular Neuronais/antagonistas & inibidores , Moléculas de Adesão Celular Neuronais/genética , Proteínas da Matriz Extracelular/antagonistas & inibidores , Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Neurogênese/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Ratos , Ratos Wistar , Proteína Reelina , Serina Endopeptidases/genética
3.
Int J Mol Sci ; 14(8): 16817-35, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23955266

RESUMO

Trimethyltin (TMT) is an organotin compound exhibiting neurotoxicant effects selectively localized in the limbic system and especially marked in the hippocampus, in both experimental animal models and accidentally exposed humans. TMT administration causes selective neuronal death involving either the granular neurons of the dentate gyrus or the pyramidal cells of the Cornu Ammonis, with a different pattern of localization depending on the different species studied or the dosage schedule. TMT is broadly used to realize experimental models of hippocampal neurodegeneration associated with cognitive impairment and temporal lobe epilepsy, though the molecular mechanisms underlying the associated selective neuronal death are still not conclusively clarified. Experimental evidence indicates that TMT-induced neurodegeneration is a complex event involving different pathogenetic mechanisms, probably acting differently in animal and cell models, which include neuroinflammation, intracellular calcium overload, and oxidative stress. Microarray-based, genome-wide expression analysis has been used to investigate the molecular scenario occurring in the TMT-injured brain in different in vivo and in vitro models, producing an overwhelming amount of data. The aim of this review is to discuss and rationalize the state-of-the-art on TMT-associated genome wide expression profiles in order to identify comparable and reproducible data that may allow focusing on significantly involved pathways.


Assuntos
Perfilação da Expressão Gênica , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Doenças Neurodegenerativas/metabolismo , Compostos de Trimetilestanho/administração & dosagem , Animais , Linhagem Celular , Camundongos , Mitocôndrias/efeitos dos fármacos , Modelos Animais , Doenças Neurodegenerativas/induzido quimicamente , Doenças Neurodegenerativas/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeos/efeitos dos fármacos , Neurotoxinas/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Ratos
4.
Transl Psychiatry ; 13(1): 114, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-37019889

RESUMO

Autism spectrum disorder (ASD) includes a set of highly heritable neurodevelopmental syndromes characterized by social and communication impairment, repetitive behaviour, and intellectual disability. Although mutations in multiple genes have been associated to ASD, most patients lack detectable genetic alterations. For this reason, environmental factors are commonly thought to also contribute to ASD aetiology. Transcriptome analyses have revealed that autistic brains possess distinct gene expression signatures, whose elucidation can provide insights about the mechanisms underlying the effects of ASD-causing genetic and environmental factors. Herein, we have identified a coordinated and temporally regulated programme of gene expression in the post-natal development of cerebellum, a brain area whose defects are strongly associated with ASD. Notably, this cerebellar developmental programme is significantly enriched in ASD-linked genes. Clustering analyses highlighted six different patterns of gene expression modulated during cerebellar development, with most of them being enriched in functional processes that are frequently dysregulated in ASD. By using the valproic acid mouse model of ASD, we found that ASD-linked genes are dysregulated in the developing cerebellum of ASD-like mice, a defect that correlates with impaired social behaviour and altered cerebellar cortical morphology. Moreover, changes in transcript levels were reflected in aberrant protein expression, indicating the functional relevance of these alterations. Thus, our work uncovers a complex ASD-related transcriptional programme regulated during cerebellar development and highlight genes whose expression is dysregulated in this brain area of an ASD mouse model.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Camundongos , Animais , Transtorno Autístico/metabolismo , Ácido Valproico , Transtorno do Espectro Autista/genética , Cerebelo/metabolismo , Encéfalo
5.
J Neurochem ; 120(5): 644-59, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22145907

RESUMO

S100B is a calcium-binding protein concentrated in glial cells, although it has also been detected in definite extra-neural cell types. Its biological role is still debated. When secreted, S100B is believed to have paracrine/autocrine trophic effects at physiological concentrations, but toxic effects at higher concentrations. Elevated S100B levels in biological fluids (CSF, blood, urine, saliva, amniotic fluid) are thus regarded as a biomarker of pathological conditions, including perinatal brain distress, acute brain injury, brain tumors, neuroinflammatory/neurodegenerative disorders, psychiatric disorders. In the majority of these conditions, high S100B levels offer an indicator of cell damage when standard diagnostic procedures are still silent. The key question remains as to whether S100B is merely leaked from injured cells or is released in concomitance with both physiological and pathological conditions, participating at high concentrations in the events leading to cell injury. In this respect, S100B levels in biological fluids have been shown to increase in physiological conditions characterized by stressful physical and mental activity, suggesting that it may be physiologically regulated and raised during conditions of stress, with a putatively active role. This possibility makes this protein a candidate not only for a biomarker but also for a potential therapeutic target.


Assuntos
Líquidos Corporais/metabolismo , Encefalopatias/patologia , Encéfalo/metabolismo , Transtornos Mentais/patologia , Fatores de Crescimento Neural/metabolismo , Proteínas S100/metabolismo , Biomarcadores/metabolismo , Humanos , Subunidade beta da Proteína Ligante de Cálcio S100
6.
J Neurochem ; 122(2): 415-26, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22537092

RESUMO

The effects of intracerebroventricular administration of neuropeptide Y (NPY), which is believed to play an important role in neuroprotection against excitotoxicity and in the modulation of adult neurogenesis, were evaluated in an animal model of hippocampal neurodegeneration and temporal lobe epilepsy represented by trimethyltin (TMT) intoxication. A single TMT injection (8 mg/kg) causes, in the rat brain, massive neuronal death, selectively involving pyramidal neurons, accompanied by glial activation and enhanced hippocampal neurogenesis. Our data indicate that intracerebroventricular administration of exogenous NPY (at the dose of 2 µg/2 µL, 4 days after TMT-administration), in adult rats, exerts a protective role in regard to TMT-induced hippocampal damage and a proliferative effect on the hippocampal neurogenic niche through the up-regulation of Bcl-2, Bcl2l1, Bdnf, Sox-2, NeuroD1, Noggin and Doublecortin genes, contributing to delineate more clearly the role of NPY in in vivo neurodegenerative processes.


Assuntos
Epilepsia do Lobo Temporal/tratamento farmacológico , Epilepsia do Lobo Temporal/patologia , Hipocampo/efeitos dos fármacos , Degeneração Neural/prevenção & controle , Neurogênese/efeitos dos fármacos , Neuropeptídeo Y/farmacologia , Fármacos Neuroprotetores , Compostos de Trimetilestanho , Animais , Antimetabólitos , Proteínas Reguladoras de Apoptose/biossíntese , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Bromodesoxiuridina , Proteína Duplacortina , Epilepsia do Lobo Temporal/induzido quimicamente , Feminino , Expressão Gênica/efeitos dos fármacos , Hipocampo/patologia , Imuno-Histoquímica , Injeções Intraventriculares , Degeneração Neural/etiologia , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Neuropeptídeo Y/administração & dosagem , RNA/biossíntese , RNA/isolamento & purificação , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Neuropeptídeo Y/efeitos dos fármacos
7.
Front Mol Neurosci ; 15: 1073627, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36710925

RESUMO

Multiple sclerosis (MS) and its preclinical models are characterized by marked changes in neuroplasticity, including excitatory/inhibitory imbalance and synaptic dysfunction that are believed to underlie the progressive cognitive impairment (CI), which represents a significant clinical hallmark of the disease. In this study, we investigated several parameters of neuroplasticity in the hippocampus of the experimental autoimmune encephalomyelitis (EAE) SJL/J mouse model, characterized by rostral inflammatory and demyelinating lesions similar to Relapsing-Remitting MS. By combining morphological and molecular analyses, we found that the hippocampus undergoes extensive inflammation in EAE-mice, more pronounced in the CA3 and dentate gyrus (DG) subfields than in the CA1, associated with changes in GABAergic circuitry, as indicated by the increased expression of the interneuron marker Parvalbumin selectively in CA3. By laser-microdissection, we investigated the impact of EAE on the alternative splicing of Arhgef9, a gene encoding a post-synaptic protein playing an essential role in GABAergic synapses and whose mutations have been related to CI and epilepsy. Our results indicate that EAE induces a specific increase in inclusion of the alternative exon 11a only in the CA3 and DG subfields, in line with the higher local levels of inflammation. Consistently, we found a region-specific downregulation of Sam68, a splicing-factor that represses this splicing event. Collectively, our findings confirm a regionalized distribution of inflammation in the hippocampus of EAE-mice. Moreover, since neuronal circuit rearrangement and dynamic remodeling of structural components of the synapse are key processes that contribute to neuroplasticity, our study suggests potential new molecular players involved in EAE-induced hippocampal dysfunction.

8.
J Biomed Biotechnol ; 2011: 468705, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22219658

RESUMO

Due to its abundance, easy retrieval, and plasticity characteristics, adipose-tissue-derived stromal cells (ATSCs) present unquestionable advantages over other adult-tissue-derived stem cells. Based on the in silico analysis of our previous data reporting the ATSC-specific expression profiles, the present study attempted to clarify and validate at the functional level the expression of the neurospecific genes expressed by ATSC both in vitro and in vivo. This allowed evidencing that ATSCs express neuro-specific trophins, metabolic genes, and neuroprotective molecules. They were in fact able to induce neurite outgrowth in vitro, along with tissue-specific commitment along the neural lineage and the expression of the TRKA neurotrophin receptor in vivo. Our observation adds useful information to recent evidence proposing these cells as a suitable tool for cell-based applications in neuroregenerative medicine.


Assuntos
Tecido Adiposo/citologia , Moléculas de Adesão Celular/metabolismo , Técnicas de Cocultura/métodos , Neuritos/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Células Estromais/metabolismo , Tecido Adiposo/metabolismo , Adulto , Animais , Diferenciação Celular , Células Cultivadas , Meios de Cultivo Condicionados , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Células PC12 , Cultura Primária de Células , Ratos , Regulação para Cima
9.
Exp Neurol ; 335: 113497, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33058888

RESUMO

Mounting evidence points to immune-mediated synaptopathy and impaired plasticity as early pathogenic events underlying cognitive decline (CD) in Multiple sclerosis (MS) and in the experimental autoimmune encephalomyelitis (EAE) mouse model of the disease. However, knowledge of the neurobiology of synaptic dysfunction is still incomplete. Splicing regulation represents a flexible and powerful mechanism involved in dynamic remodeling of the synapse, which allows the expression of synaptic protein variants that dynamically control the specificity of contacts between neurons. The pre-synaptic adhesion molecules neurexins (NRXNs) 1-3 play a relevant role in cognition and are alternatively spliced to yield variants that differentially cluster specific ligands in the postsynaptic compartment and modulate functional properties of the synaptic contact. Notably, mutations in these genes or disruption of their splicing program are associated with neuropsychiatric disorders. Herein, we have investigated how inflammatory changes imposed by EAE impact on alternative splicing of the Nrxn 1-3 mouse genes in the acute phase of disease. Due to its relevance in cognition, we focused on the prefrontal cortex (PFC) of SJL/J mice, in which EAE-induced inflammatory lesions extend to the rostral forebrain. We found that inclusion of the Nrxn 1-3 AS4 exon is significantly increased in the PFC of EAE mice and that splicing changes are correlated with local Il1ß-expression levels. This correlation is sustained by the concomitant downregulation of SLM2, the main splicing factor involved in skipping of the AS4 exon, in EAE mice displaying high levels of Il1ß- expression. We also observed that Il1ß-expression levels correlate with changes in parvalbumin (PV)-positive interneuron connectivity. Moreover, exposure to environmental enrichment (EE), a condition known to stimulate neuronal connectivity and to improve cognitive functions in mice and humans, modified PFC phenotypes of EAE mice with respect to Il1ß-, Slm2-expression, Nrxn AS4 splicing and PV-expression, by limiting changes associated with high levels of inflammation. Our results reveal that local inflammation results in early splicing modulation of key synaptic proteins and in remodeling of GABAergic circuitry in the PFC of SJL/J mice. We also suggest EE as a tool to counteract these inflammation-associated events, thus highlighting potential therapeutic targets for limiting the progressive CD occurring in MS.


Assuntos
Processamento Alternativo/genética , Proteínas de Ligação ao Cálcio/genética , Disfunção Cognitiva/genética , Disfunção Cognitiva/psicologia , Encefalite/genética , Encefalite/psicologia , Esclerose Múltipla/genética , Esclerose Múltipla/psicologia , Proteínas do Tecido Nervoso/genética , Moléculas de Adesão de Célula Nervosa/genética , Córtex Pré-Frontal/patologia , Animais , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/psicologia , Éxons/genética , Feminino , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Interneurônios , Camundongos , Vias Neurais , Reconhecimento Psicológico , Ácido gama-Aminobutírico
10.
J Neurosci Res ; 88(3): 500-9, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19795376

RESUMO

Nitric oxide (NO) and purinergic ionotropic receptors (P2X) mediate cellular events in the central nervous system (CNS) under physiological conditions as well as during pathological events, and they have been recently proposed to interact in mediating CNS response to injury (Viscomi et al. [2004] Neuroscience 123:393-404; Florenzano et al. [2008] Pflugers Arch. 452:622-644). Trimethyltin (TMT) is an organotin compound that generates neurotoxic effects, and it has been used in a model of neurodegenerative disease and memory dysfunction. TMT causes neuronal death and reactive gliosis primarily in the hippocampus and other limbic regions. In the present study, we examined the degenerative events and the expression of nitric oxide synthase (NOS) and P2X receptor subtypes (P2X(1,2,4,7)Rs) that were induced by TMT administration at different time points (3, 7, 14, and 21 days) by conventional and confocal microscopy and Western blotting. Massive glial activation and neuronal death in the CA1 and CA3 regions were observed after TMT treatment. In these areas, astrocytic P2X(2)R and neuronal NOS were temporarily enhanced in association with the progression of neuronal death. In the hippocampus, the physiological expression of P2X(1)R, P2X(4)R, and P2X(7)R was not modified by TMT. The present data demonstrate that, as in other neurodegenerative models, TMT-induced hippocampal degeneration is associated with nitrergic and purinergic activations. Nevertheless, at odds with previous data, in this model the two systems are active in segregated cell populations, namely, P2XR in astrocytes and NOS in neurons. Finally, the temporal relations between P2XR and NOS expression and neuronal degeneration suggest interactions between P2XR/NO signaling and cell survival.


Assuntos
Fármacos do Sistema Nervoso Central/toxicidade , Hipocampo/efeitos dos fármacos , Óxido Nítrico Sintase/metabolismo , Receptores Purinérgicos P2/metabolismo , Compostos de Trimetilestanho/toxicidade , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/enzimologia , Astrócitos/fisiologia , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/enzimologia , Região CA1 Hipocampal/fisiologia , Região CA3 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/enzimologia , Região CA3 Hipocampal/fisiologia , Morte Celular/efeitos dos fármacos , Feminino , Hipocampo/enzimologia , Hipocampo/fisiologia , Degeneração Neural/induzido quimicamente , Degeneração Neural/enzimologia , Degeneração Neural/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/fisiologia , Óxido Nítrico Sintase Tipo I/metabolismo , Ratos , Ratos Wistar , Receptores Purinérgicos P2X , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos
11.
Front Cell Neurosci ; 12: 385, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30416427

RESUMO

Hippocampal dysfunction plays a central role in neurodevelopmental disorders, resulting in severe impairment of cognitive abilities, including memory and learning. On this basis, developmental studies represent an important tool both to understanding the cellular and molecular phenomena underlying early hippocampal damage and to study possible therapeutic interventions, that may modify the progression of neuronal death. Given the modulatory role played by 17ß-estradiol (E2) on hippocampal functions and its neuroprotective properties, the present study investigates the effects of pretreatment with E2 in a model of neonatal hippocampal injury obtained by trimethyltin (TMT) administration, characterized by neuronal loss in CA1 and CA3 subfields and astroglial and microglial activation. At post-natal days (P)5 and P6 animals received E2 administration (0.2 mg/kg/die i.p.) or vehicle. At P7 they received a single dose of TMT (6.5 mg/kg i.p.) and were sacrificed 72 h (P10) or 7 days after TMT treatment (P14). Our findings indicate that pretreatment with E2 exerts a protective effect against hippocampal damage induced by TMT administration early in development, reducing the extent of neuronal death in the CA1 subfield, inducing the activation of genes involved in neuroprotection, lowering the neuroinflammatory response and restoring neuropeptide Y- and parvalbumin- expression, which is impaired in the early phases of TMT-induced damage. Our data support the efficacy of estrogen-based neuroprotective approaches to counteract early occurring hippocampal damage in the developing hippocampus.

12.
Front Aging Neurosci ; 9: 242, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28790913

RESUMO

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by a non-cell autonomous motor neuron loss. While it is generally believed that the disease onset takes place inside motor neurons, different cell types mediating neuroinflammatory processes are considered deeply involved in the progression of the disease. On these grounds, many treatments have been tested on ALS animals with the aim of inhibiting or reducing the pro-inflammatory action of microglia and astrocytes and counteract the progression of the disease. Unfortunately, these anti-inflammatory therapies have been only modestly successful. The non-univocal role played by microglia during stress and injuries might explain this failure. Indeed, it is now well recognized that, during ALS, microglia displays different phenotypes, from surveillant in early stages, to activated states, M1 and M2, characterized by the expression of respectively harmful and protective genes in later phases of the disease. Consistently, the inhibition of microglial function seems to be a valid strategy only if the different stages of microglia polarization are taken into account, interfering with the reactivity of microglia specifically targeting only the harmful pathways and/or potentiating the trophic ones. In this review article, we will analyze the features and timing of microglia activation in the light of M1/M2 phenotypes in the main mice models of ALS. Moreover, we will also revise the results obtained by different anti-inflammatory therapies aimed to unbalance the M1/M2 ratio, shifting it towards a protective outcome.

13.
Biochim Biophys Acta ; 1619(2): 209-12, 2003 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-12527118

RESUMO

The present study constitutes the first finding of the calcium-binding protein S100B and of its mRNA in human milk, as revealed by a quantitative immunoluminometric assay, by Western blot analysis and by reverse transcription-polymerase chain reaction (RT-PCR) assay followed by restriction enzyme digestion. The concentration of S100B in milk is markedly higher than that observed in other biological fluids such as cord blood, peripheral blood, urine, cerebrospinal fluid and amniotic fluid. This finding could be related to a possible trophic role, which has been hypothesized for the protein.


Assuntos
Leite Humano/química , Fatores de Crescimento Neural/análise , Proteínas S100/análise , Western Blotting , Feminino , Humanos , Medições Luminescentes , Fatores de Crescimento Neural/química , RNA Mensageiro/análise , Mapeamento por Restrição , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/química
14.
Clin Biochem ; 38(3): 229-33, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15708543

RESUMO

OBJECTIVES: S100B is an acidic calcium-binding protein of the EF-hand family present in the central nervous system, where it is concentrated in glial cells. It has been suggested to act as a cytokine with neurotrophic effects at physiological concentrations. DESIGN AND METHODS: S100B concentration was assessed in saliva by western blot analysis and an immunoluminometric assay. A reference curve of the protein was established in 216 preterm and term newborns. RESULTS: S100B levels were significantly higher in saliva taken from the preterm group, and the highest S100B levels were found in newborns who were delivered in the earlier weeks of gestation, exhibiting a progressive decrease nearer to term. S100B concentration in saliva was correlated with gestational age (r = -0.69; P < 0.001). CONCLUSIONS: The present study offers data consistent with the putative neurotrophic role of S100B and suggests the usefulness of saliva in the clinical monitoring of S100B levels.


Assuntos
Biomarcadores/análise , Idade Gestacional , Recém-Nascido/metabolismo , Recém-Nascido Prematuro/metabolismo , Proteínas S100/análise , Saliva/química , Western Blotting , Feminino , Humanos , Masculino , Fatores de Crescimento Neural , Valores de Referência , Subunidade beta da Proteína Ligante de Cálcio S100
15.
Brain Res Bull ; 65(6): 471-7, 2005 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-15862918

RESUMO

The occurrence of neurogenesis in the hippocampus of the adult rat during trimethyltin (TMT)-induced neurodegeneration was investigated using bromodeoxyuridine (BrdU). Fifteen days after TMT intoxication, BrdU-labeled cells were significantly more numerous in the hippocampus of treated animals, gradually decreasing towards the control value 21 days after intoxication in the dentate gyrus (DG), while in the CA3/hilus region BrdU-labeled cells were still more numerous in TMT-treated rats. In order to investigate the fate of newly-generated cells double labeling experiments using neuronal or glial markers were performed. Colocalization of the neuronal marker NeuN was detected in many BrdU-positive cells in the DG, while in the CA3/hilus region no colocalization of NeuN and BrdU could be observed. No colocalization of BrdU and the astroglial marker GFAP or the microglial marker OX-42 was detected either in the DG and or in the CA3/hilus region. The results indicate an enhancement of endogenous neurogenesis in the hippocampus during TMT-induced neurodegeneration, with the development of a subpopulation of regenerated cells into neurons in the DG, while in the CA3/hilus region the population of newly-generated cells should be regarded as undifferentiated.


Assuntos
Hipocampo/patologia , Degeneração Neural/fisiopatologia , Neurônios/fisiologia , Regeneração/fisiologia , Animais , Bromodesoxiuridina/metabolismo , Antígeno CD11b/metabolismo , Contagem de Células/métodos , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica/métodos , Masculino , Degeneração Neural/induzido quimicamente , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Wistar , Coloração e Rotulagem/métodos , Compostos de Trimetilestanho/metabolismo
16.
Front Cell Neurosci ; 9: 433, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26594149

RESUMO

Given the well-documented involvement of estrogens in the modulation of hippocampal functions in both physiological and pathological conditions, the present study investigates the effects of 17-beta estradiol (E2) administration in the rat model of hippocampal neurodegeneration induced by trimethyltin (TMT) administration (8 mg/kg), characterized by loss of pyramidal neurons in CA1, CA3/hilus hippocampal subfields, associated with astroglial and microglial activation, seizures and cognitive impairment. After TMT/saline treatment, ovariectomized animals received two doses of E2 (0.2 mg/kg intra-peritoneal) or vehicle, and were sacrificed 48 h or 7 days after TMT-treatment. Our results indicate that in TMT-treated animals E2 administration induces the early (48 h) upregulation of genes involved in neuroprotection and synaptogenesis, namely Bcl2, trkB, cadherin 2 and cyclin-dependent-kinase-5. Increased expression levels of glutamic acid decarboxylase (gad) 67, neuropeptide Y (Npy), parvalbumin, Pgc-1α and Sirtuin 1 genes, the latter involved in parvalbumin (PV) synthesis, were also evident. Unbiased stereology performed on rats sacrificed 7 days after TMT treatment showed that although E2 does not significantly influence the extent of TMT-induced neuronal death, significantly enhances the TMT-induced modulation of GABAergic interneuron population size in selected hippocampal subfields. In particular, E2 administration causes, in TMT-treated rats, a significant increase in the number of GAD67-expressing interneurons in CA1 stratum oriens, CA3 pyramidal layer, hilus and dentate gyrus, accompanied by a parallel increase in NPY-expressing cells, essentially in the same regions, and of PV-positive cells in CA1 pyramidal layer. The present results add information concerning the role of in vivo E2 administration on mechanisms involved in cellular plasticity in the adult brain.

17.
Front Cell Neurosci ; 9: 85, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25852477

RESUMO

Neuropeptides are emerging as key regulators of stem cell niche activities in health and disease, both inside and outside the central nervous system (CNS). Among them, neuropeptide Y (NPY), one of the most abundant neuropeptides both in the nervous system and in non-neural districts, has become the focus of much attention for its involvement in a wide range of physiological and pathological conditions, including the modulation of different stem cell activities. In particular, a pro-neurogenic role of NPY has been evidenced in the neurogenic niche, where a direct effect on neural progenitors has been demonstrated, while different cellular types, including astrocytes, microglia and endothelial cells, also appear to be responsive to the peptide. The marked modulation of the NPY system during several pathological conditions that affect neurogenesis, including stress, seizures and neurodegeneration, further highlights the relevance of this peptide in the regulation of adult neurogenesis. In view of the considerable interest in understanding the mechanisms controlling neural cell fate, this review aims to summarize and discuss current data on NPY signaling in the different cellular components of the neurogenic niche in order to elucidate the complexity of the mechanisms underlying the modulatory properties of this peptide.

18.
Brain Res Mol Brain Res ; 122(1): 93-8, 2004 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-14992820

RESUMO

Thrombin, its main inhibitor (protease nexin-1) and its related receptors (protease-activated receptors, PAR-1,-2, -3, -4) were studied in rat hippocampus following administration of trimethyltin (TMT), a neurotoxin inducing neuronal degeneration and reactive gliosis. Reverse transcriptase-polymerase chain reaction (RT-PCR) and immunohistochemistry revealed that while expression of prothrombin and protease nexin-1 did not change significantly in TMT-treated hippocampi, PARs (in particular PAR-1 and to a lesser extent PAR-2 and PAR-3) were upregulated in reactive astrocytes, suggesting their involvement in neurodegeneration and in the consequent response of the nervous tissue.


Assuntos
Astrócitos/efeitos dos fármacos , Hipocampo/citologia , Neurotoxinas/farmacologia , Receptores Ativados por Proteinase/metabolismo , Compostos de Trimetilestanho/farmacologia , Animais , Astrócitos/metabolismo , Hipocampo/efeitos dos fármacos , Imuno-Histoquímica/métodos , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Receptores Ativados por Proteinase/classificação , Receptores Ativados por Proteinase/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Regulação para Cima
19.
Brain Res Mol Brain Res ; 98(1-2): 141-4, 2002 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-11834306

RESUMO

In the model of trimethyltin (TMT)-induced neurodegeneration in developing rat hippocampus, calretinin (CR)-immunoreactive neurons are selectively spared and even more numerous than in controls. We investigated the possibility of an additional synthesis of CR using RT-PCR. The amount of CR mRNA increased significantly after TMT treatment. CR mRNA production after TMT treatment could hypothetically be regarded as a compensatory phenomenon in developing rats.


Assuntos
Hipocampo/efeitos dos fármacos , Degeneração Neural/induzido quimicamente , Proteínas do Tecido Nervoso/biossíntese , Neurotoxinas/toxicidade , Proteína G de Ligação ao Cálcio S100/biossíntese , Compostos de Trimetilestanho/toxicidade , Animais , Calbindina 2 , Cálcio/metabolismo , DNA Complementar/genética , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Degeneração Neural/genética , Degeneração Neural/metabolismo , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína G de Ligação ao Cálcio S100/genética
20.
Obstet Gynecol ; 99(6): 1093-9, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12052605

RESUMO

OBJECTIVE: To investigate whether S-100beta, a brain-specific protein found in amniotic fluid and fetal circulation, is present in fetoplacental tissues throughout gestation. METHODS: S-100beta protein localization and concentration were assessed in placentae, fetal membranes, and cord vessels. Tissues were obtained from 40 pregnant women at different gestational ages: first trimester (n = 10), second trimester (n = 10), early third trimester (n = 10), and late third trimester (n = 10). RESULTS: In the placenta, S-100beta was localized in villous and intermediate trophoblast cells. The intensity of immunostaining and protein concentration increased with advancing gestation. S-100beta protein was also present in amnion, trophoblast, and decidual cells of fetal membranes, and in endothelial cells of umbilical vessels at all gestational ages. CONCLUSION: This study demonstrated that fetoplacental tissues contain S-100beta protein, suggesting that these tissues may, at least in part, be responsible for the high level found in the fetal circulation. Although the significance of placental S-100beta is unknown, this origin should be taken into account when this protein is used as a marker of brain injury in the fetus or infant at birth.


Assuntos
Membranas Extraembrionárias/metabolismo , Placenta/metabolismo , Gravidez/metabolismo , Proteínas S100/metabolismo , Veias Umbilicais/metabolismo , Western Blotting , Feminino , Idade Gestacional , Humanos , Imuno-Histoquímica , Fatores de Crescimento Neural , Trimestres da Gravidez , Subunidade beta da Proteína Ligante de Cálcio S100 , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA