Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Glia ; 72(5): 899-915, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38288580

RESUMO

Alzheimer's disease (AD) represents an urgent yet unmet challenge for modern society, calling for exploration of innovative targets and therapeutic approaches. Astrocytes, main homeostatic cells in the CNS, represent promising cell-target. Our aim was to investigate if deletion of the regulatory CaNB1 subunit of calcineurin in astrocytes could mitigate AD-related memory deficits, neuropathology, and neuroinflammation. We have generated two, acute and chronic, AD mouse models with astrocytic CaNB1 ablation (ACN-KO). In the former, we evaluated the ability of ß-amyloid oligomers (AßOs) to impair memory and activate glial cells once injected in the cerebral ventricle of conditional ACN-KO mice. Next, we generated a tamoxifen-inducible astrocyte-specific CaNB1 knock-out in 3xTg-AD mice (indACNKO-AD). CaNB1 was deleted, by tamoxifen injection, in 11.7-month-old 3xTg-AD mice for 4.4 months. Spatial memory was evaluated using the Barnes maze; ß-amyloid plaques burden, neurofibrillary tangle deposition, reactive gliosis, and neuroinflammation were also assessed. The acute model showed that ICV injected AßOs in 2-month-old wild type mice impaired recognition memory and fostered a pro-inflammatory microglia phenotype, whereas in ACN-KO mice, AßOs were inactive. In indACNKO-AD mice, 4.4 months after CaNB1 depletion, we found preservation of spatial memory and cognitive flexibility, abolishment of amyloidosis, and reduction of neurofibrillary tangles, gliosis, and neuroinflammation. Our results suggest that ACN is crucial for the development of cognitive impairment, AD neuropathology, and neuroinflammation. Astrocyte-specific CaNB1 deletion is beneficial for both the abolishment of AßO-mediated detrimental effects and treatment of ongoing AD-related pathology, hence representing an intriguing target for AD therapy.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Camundongos , Animais , Doença de Alzheimer/patologia , Astrócitos/patologia , Calcineurina , Gliose/patologia , Doenças Neuroinflamatórias , Peptídeos beta-Amiloides , Disfunção Cognitiva/genética , Disfunção Cognitiva/patologia , Tamoxifeno/farmacologia , Modelos Animais de Doenças , Camundongos Transgênicos , Camundongos Endogâmicos C57BL
2.
Acta Neuropathol ; 147(1): 48, 2024 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-38418708

RESUMO

Tuberous Sclerosis Complex (TSC) is a multisystem genetic disorder characterized by the development of benign tumors in various organs, including the brain, and is often accompanied by epilepsy, neurodevelopmental comorbidities including intellectual disability and autism. A key hallmark of TSC is the hyperactivation of the mechanistic target of rapamycin (mTOR) signaling pathway, which induces alterations in cortical development and metabolic processes in astrocytes, among other cellular functions. These changes could modulate seizure susceptibility, contributing to the progression of epilepsy and its associated comorbidities. Epilepsy is characterized by dysregulation of calcium (Ca2+) channels and intracellular Ca2+ dynamics. These factors contribute to hyperexcitability, disrupted synaptogenesis, and altered synchronization of neuronal networks, all of which contribute to seizure activity. This study investigates the intricate interplay between altered Ca2+ dynamics, mTOR pathway dysregulation, and cellular metabolism in astrocytes. The transcriptional profile of TSC patients revealed significant alterations in pathways associated with cellular respiration, ER and mitochondria, and Ca2+ regulation. TSC astrocytes exhibited lack of responsiveness to various stimuli, compromised oxygen consumption rate and reserve respiratory capacity underscoring their reduced capacity to react to environmental changes or cellular stress. Furthermore, our study revealed significant reduction of store operated calcium entry (SOCE) along with strong decrease of basal mitochondrial Ca2+ concentration and Ca2+ influx in TSC astrocytes. In addition, we observed alteration in mitochondrial membrane potential, characterized by increased depolarization in TSC astrocytes. Lastly, we provide initial evidence of structural abnormalities in mitochondria within TSC patient-derived astrocytes, suggesting a potential link between disrupted Ca2+ signaling and mitochondrial dysfunction. Our findings underscore the complexity of the relationship between Ca2+ signaling, mitochondria dynamics, apoptosis, and mTOR hyperactivation. Further exploration is required to shed light on the pathophysiology of TSC and on TSC associated neuropsychiatric disorders offering further potential avenues for therapeutic development.


Assuntos
Epilepsia , Esclerose Tuberosa , Humanos , Astrócitos/patologia , Sinalização do Cálcio , Esclerose Tuberosa/patologia , Cálcio/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Epilepsia/genética , Homeostase , Convulsões
3.
Neurochem Res ; 48(4): 1077-1090, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36083398

RESUMO

Calcineurin (CaN), a Ca2+/calmodulin-activated serine/threonine phosphatase, acts as a Ca2+-sensitive switch regulating cellular functions through protein dephosphorylation and activation of gene transcription. In astrocytes, the principal homeostatic cells in the CNS, over-activation of CaN is known to drive pathological transcriptional remodelling, associated with neuroinflammation in diseases such as Alzheimer's disease, epilepsy and brain trauma. Recent reports suggest that, in physiological conditions, the activity of CaN in astrocytes is transcription-independent and is required for maintenance of basal protein synthesis rate and activation of astrocytic Na+/K+ pump thereby contributing to neuronal functions such as neuronal excitability and memory formation. In this contribution we overview the role of Ca2+ and CaN signalling in astroglial pathophysiology focusing on the emerging physiological role of CaN in astrocytes. We propose a model for the context-dependent switch of CaN activity from the post-transcriptional regulation of cell proteostasis in healthy astrocytes to the CaN-dependent transcriptional activation in neuroinflammation-associated diseases.


Assuntos
Doença de Alzheimer , Astrócitos , Humanos , Astrócitos/metabolismo , Calcineurina/metabolismo , Doenças Neuroinflamatórias , Neurônios/metabolismo , Doença de Alzheimer/metabolismo
4.
Blood ; 135(2): 133-144, 2020 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-31697806

RESUMO

Approximately one-fourth of patients with essential thrombocythemia or primary myelofibrosis carry a somatic mutation of the calreticulin gene (CALR), the gene encoding for calreticulin. A 52-bp deletion (type I mutation) and a 5-bp insertion (type II mutation) are the most frequent genetic lesions. The mechanism(s) by which a CALR mutation leads to a myeloproliferative phenotype has been clarified only in part. We studied the interaction between calreticulin and store-operated calcium (Ca2+) entry (SOCE) machinery in megakaryocytes (Mks) from healthy individuals and from patients with CALR-mutated myeloproliferative neoplasms (MPNs). In Mks from healthy subjects, binding of recombinant human thrombopoietin to c-Mpl induced the activation of signal transducer and activator of transcription 5, AKT, and extracellular signal-regulated kinase 1/2, determining inositol triphosphate-dependent Ca2+ release from the endoplasmic reticulum (ER). This resulted in the dissociation of the ER protein 57 (ERp57)-mediated complex between calreticulin and stromal interaction molecule 1 (STIM1), a protein of the SOCE machinery that leads to Ca2+ mobilization. In Mks from patients with CALR-mutated MPNs, defective interactions between mutant calreticulin, ERp57, and STIM1 activated SOCE and generated spontaneous cytosolic Ca2+ flows. In turn, this resulted in abnormal Mk proliferation that was reverted using a specific SOCE inhibitor. In summary, the abnormal SOCE regulation of Ca2+ flows in Mks contributes to the pathophysiology of CALR-mutated MPNs. In perspective, SOCE may represent a new therapeutic target to counteract Mk proliferation and its clinical consequences in MPNs.


Assuntos
Canais de Cálcio Ativados pela Liberação de Cálcio/metabolismo , Calreticulina/genética , Calreticulina/metabolismo , Megacariócitos/patologia , Mutação , Transtornos Mieloproliferativos/patologia , Canais de Cálcio Ativados pela Liberação de Cálcio/genética , Estudos de Casos e Controles , Humanos , Megacariócitos/metabolismo , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo
5.
Cell Mol Neurobiol ; 41(3): 551-562, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32440709

RESUMO

Impairments of the blood-brain barrier (BBB) and vascular dysfunction contribute to Alzheimer's disease (AD) from the earliest stages. However, the influence of AD-affected astrocytes on the BBB remain largely unexplored. In the present study, we created an in vitro BBB using human-immortalized endothelial cells in combination with immortalized astroglial cell lines from the hippocampus of 3xTG-AD and wild-type mice (3Tg-iAstro and WT-iAstro, respectively). We found that co-culturing endothelial monolayers with WT-iAstro upregulates expression of endothelial tight junction proteins (claudin-5, occludin, ZO-1) and increases the trans-endothelial electrical resistance (TEER). In contrast, co-culturing with 3Tg-iAstro does not affect expression of tight junction proteins and does not change the TEER of endothelial monolayers. The same in vitro model has been used to evaluate the effects of extracellular vesicles (EVs) derived from the WT-iAstro and 3Tg-iAstro. The EVs derived from WT-iAstro increased TEER and upregulated expression of tight junction proteins, whereas EVs from 3Tg-iAstro were ineffective. In conclusion, we show for the first time that immortalized hippocampal astrocytes from 3xTG-AD mice exhibit impaired capacity to support BBB integrity in vitro through paracrine mechanisms and may represent an important factor underlying vascular abnormalities during development of AD.


Assuntos
Doença de Alzheimer/patologia , Astrócitos/metabolismo , Barreira Hematoencefálica/patologia , Comunicação Celular , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Hipocampo/patologia , Neuroglia/metabolismo , Doença de Alzheimer/genética , Animais , Astrócitos/efeitos dos fármacos , Comunicação Celular/efeitos dos fármacos , Linhagem Celular Transformada , Meios de Cultivo Condicionados/farmacologia , Impedância Elétrica , Células Endoteliais/efeitos dos fármacos , Vesículas Extracelulares/efeitos dos fármacos , Vesículas Extracelulares/ultraestrutura , Humanos , Camundongos Transgênicos , Neuroglia/efeitos dos fármacos , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo , Regulação para Cima/genética
6.
Cell Mol Neurobiol ; 41(3): 563-587, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32430779

RESUMO

Cisplatin (CDDP) is one of the most effective chemotherapeutic agents, used for the treatment of diverse tumors, including neuroblastoma and glioblastoma. CDDP induces cell death through different apoptotic pathways. Despite its clinical benefits, CDDP causes several side effects and drug resistance.[Pt(O,O'-acac)(γ-acac)(DMS)], namely PtAcacDMS, a new platinum(II) complex containing two acetylacetonate (acac) and a dimethylsulphide (DMS) in the coordination sphere of metal, has been recently synthesized and showed 100 times higher cytotoxicity than CDDP. Additionally, PtAcacDMS was associated to a decreased neurotoxicity in developing rat central nervous system, also displaying great antitumor and antiangiogenic activity both in vivo and in vitro. Thus, based on the knowledge that several chemotherapeutics induce cancer cell death through an aberrant increase in [Ca2+]i, in the present in vitro study we compared CDDP and PtAcacDMS effects on apoptosis and intracellular Ca2+ dynamics in human glioblastoma T98G cells, applying a battery of complementary techniques, i.e., flow cytometry, immunocytochemistry, electron microscopy, Western blotting, qRT-PCR, and epifluorescent Ca2+ imaging. The results confirmed that (i) platinum compounds may induce cell death through an aberrant increase in [Ca2+]i and (ii) PtAcacDMS exerted stronger cytotoxic effect than CDDP, associated to a larger increase in resting [Ca2+]i. These findings corroborate the use of PtAcacDMS as a promising approach to improve Pt-based chemotherapy against gliomas, either by inducing a chemosensitization or reducing chemoresistance in cell lineages resilient to CDDP treatment.


Assuntos
Neoplasias Encefálicas/patologia , Cisplatino/efeitos adversos , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Glioma/patologia , Compostos Organoplatínicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/ultraestrutura , Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/genética , Glioma/ultraestrutura , Homeostase/efeitos dos fármacos , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
7.
J Biol Chem ; 294(33): 12472-12482, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31248983

RESUMO

Type 2 taste receptors (TAS2R) are G protein-coupled receptors first described in the gustatory system, but have also been shown to have extraoral localizations, including airway smooth muscle (ASM) cells, in which TAS2R have been reported to induce relaxation. TAS2R46 is an unexplored subtype that responds to its highly specific agonist absinthin. Here, we first demonstrate that, unlike other bitter-taste receptor agonists, absinthin alone (1 µm) in ASM cells does not induce Ca2+ signals but reduces histamine-induced cytosolic Ca2+ increases. To investigate this mechanism, we introduced into ASM cells aequorin-based Ca2+ probes targeted to the cytosol, subplasma membrane domain, or the mitochondrial matrix. We show that absinthin reduces cytosolic histamine-induced Ca2+ rises and simultaneously increases Ca2+ influx into mitochondria. We found that this effect is inhibited by the potent human TAS2R46 (hTAS2R46) antagonist 3ß-hydroxydihydrocostunolide and is no longer evident in hTAS2R46-silenced ASM cells, indicating that it is hTAS2R46-dependent. Furthermore, these changes were sensitive to the mitochondrial uncoupler carbonyl cyanide p-(trifluoromethoxy)phenyl-hydrazone (FCCP); the mitochondrial calcium uniporter inhibitor KB-R7943 (carbamimidothioic acid); the cytoskeletal disrupter latrunculin; and an inhibitor of the exchange protein directly activated by cAMP (EPAC), ESI-09. Similarly, the ß2 agonist salbutamol also could induce Ca2+ shuttling from cytoplasm to mitochondria, suggesting that this new mechanism might be generalizable. Moreover, forskolin and an EPAC activator mimicked this effect in HeLa cells. Our findings support the hypothesis that plasma membrane receptors can positively regulate mitochondrial Ca2+ uptake, adding a further facet to the ability of cells to encode complex Ca2+ signals.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Mitocôndrias/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Sistema Respiratório/metabolismo , Sesquiterpenos de Guaiano/farmacologia , Cálcio/metabolismo , Carbonil Cianeto p-Trifluormetoxifenil Hidrazona/farmacologia , Linhagem Celular , Retículo Endoplasmático/genética , Células HeLa , Humanos , Mitocôndrias/genética , Miócitos de Músculo Liso/citologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Sistema Respiratório/citologia , Tioureia/análogos & derivados , Tioureia/farmacologia
8.
Glia ; 68(3): 543-560, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31626368

RESUMO

Astrocytes perform important housekeeping functions in the nervous system including maintenance of adequate neuronal excitability, although the regulatory mechanisms are currently poorly understood. The astrocytic Ca2+ /calmodulin-activated phosphatase calcineurin (CaN) is implicated in the development of reactive gliosis and neuroinflammation, but its roles, including the control of neuronal excitability, in healthy brain is unknown. We have generated a mouse line with conditional knockout (KO) of CaN B1 (CaNB1) in glial fibrillary acidic protein-expressing astrocytes (astroglial calcineurin KO [ACN-KO]). Here, we report that postnatal and astrocyte-specific ablation of CaNB1 did not alter normal growth and development as well as adult neurogenesis. Yet, we found that specific deletion of astrocytic CaN selectively impairs intrinsic neuronal excitability in hippocampal CA1 pyramidal neurons and cerebellar granule cells (CGCs). This impairment was associated with a decrease in after hyperpolarization in CGC, while passive properties were unchanged, suggesting impairment of K+ homeostasis. Indeed, blockade of Na+ /K+ -ATPase (NKA) with ouabain phenocopied the electrophysiological alterations observed in ACN-KO CGCs. In addition, NKA activity was significantly lower in cerebellar and hippocampal lysates and in pure astrocytic cultures from ACN-KO mice. While no changes were found in protein levels, NKA activity was inhibited by the specific CaN inhibitor FK506 in both cerebellar lysates and primary astroglia from control mice, suggesting that CaN directly modulates NKA activity and in this manner controls neuronal excitability. In summary, our data provide formal evidence for the notion that astroglia is fundamental for controlling basic neuronal functions and place CaN center-stage as an astrocytic Ca2+ -sensitive switch.


Assuntos
Astrócitos/metabolismo , Calcineurina/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/citologia , Animais , Células Cultivadas , Cerebelo/metabolismo , Gliose/metabolismo , Camundongos Knockout , Neurônios/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
9.
Int J Mol Sci ; 21(6)2020 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-32210081

RESUMO

Alterations in the expression of glutamate/aspartate transporter (GLAST) have been associated with several neuropathological conditions including Alzheimer's disease and epilepsy. However, the mechanisms by which GLAST expression is altered are poorly understood. Here we used a combination of pharmacological and genetic approaches coupled with quantitative PCR and Western blot to investigate the mechanism of the regulation of GLAST expression by a Ca2+/calmodulin-activated phosphatase calcineurin (CaN). We show that treatment of cultured hippocampal mouse and fetal human astrocytes with a CaN inhibitor FK506 resulted in a dynamic modulation of GLAST protein expression, being downregulated after 24-48 h, but upregulated after 7 days of continuous FK506 (200 nM) treatment. Protein synthesis, as assessed by puromycin incorporation in neo-synthesized polypeptides, was inhibited already after 1 h of FK506 treatment, while the use of a proteasome inhibitor MG132 (1 µM) shows that GLAST protein degradation was only suppressed after 7 days of FK506 treatment. In astrocytes with constitutive genetic ablation of CaN both protein synthesis and degradation were significantly inhibited. Taken together, our data suggest that, in cultured astrocytes, CaN controls GLAST expression at a posttranscriptional level through regulation of GLAST protein synthesis and degradation.


Assuntos
Astrócitos/metabolismo , Calcineurina/metabolismo , Transportador 1 de Aminoácido Excitatório/genética , Regulação da Expressão Gênica , Animais , Calcineurina/farmacologia , Inibidores de Calcineurina , Células Cultivadas , Transportador 1 de Aminoácido Excitatório/metabolismo , Imunofluorescência , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Inativação de Genes , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Knockout , Modelos Moleculares , Biossíntese de Proteínas , Proteólise
10.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-32998392

RESUMO

Oxaliplatin-induced peripheral neuropathy is characterized by an acute hyperexcitability syndrome triggered/exacerbated by cold. The mechanisms underlying oxaliplatin-induced peripheral neuropathy are unclear, but the alteration of ion channel expression and activity plays a well-recognized central role. Recently, we found that oxaliplatin leads to cytosolic acidification in dorsal root ganglion (DRG) neurons. Here, we investigated the early impact of oxaliplatin on the proton-sensitive TREK potassium channels. Following a 6-h oxaliplatin treatment, both channels underwent a transcription upregulation that returned to control levels after 42 h. The overexpression of TREK channels was also observed after in vivo treatment in DRG cells from mice exposed to acute treatment with oxaliplatin. Moreover, both intracellular pH and TREK channel transcription were similarly regulated after incubation with amiloride, an inhibitor of the Na+/H+ exchanger. In addition, we studied the role of oxaliplatin-induced acidification on channel behavior, and, as expected, we observed a robust positive modulation of TREK channel activity. Finally, we focused on the impact of this complex modulation on capsaicin-evoked neuronal activity finding a transient decrease in the average firing rate following 6 h of oxaliplatin treatment. In conclusion, the early activation of TREK genes may represent a mechanism of protection against the oxaliplatin-related perturbation of neuronal excitability.


Assuntos
Antineoplásicos/efeitos adversos , Gânglios Espinais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Oxaliplatina/efeitos adversos , Doenças do Sistema Nervoso Periférico/genética , Canais de Potássio de Domínios Poros em Tandem/genética , Trocador 1 de Sódio-Hidrogênio/genética , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Amilorida/farmacologia , Animais , Capsaicina/farmacologia , Bloqueadores do Canal de Sódio Epitelial/farmacologia , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Humanos , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Neurônios/metabolismo , Neurônios/patologia , Técnicas de Patch-Clamp , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/patologia , Canais de Potássio de Domínios Poros em Tandem/agonistas , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Cultura Primária de Células , Trocador 1 de Sódio-Hidrogênio/antagonistas & inibidores , Trocador 1 de Sódio-Hidrogênio/metabolismo , Ativação Transcricional
11.
J Cell Physiol ; 234(4): 3538-3554, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30451297

RESUMO

The neurotransmitter glutamate increases cerebral blood flow by activating postsynaptic neurons and presynaptic glial cells within the neurovascular unit. Glutamate does so by causing an increase in intracellular Ca2+ concentration ([Ca2+ ]i ) in the target cells, which activates the Ca2+ /Calmodulin-dependent nitric oxide (NO) synthase to release NO. It is unclear whether brain endothelial cells also sense glutamate through an elevation in [Ca2+ ]i and NO production. The current study assessed whether and how glutamate drives Ca2+ -dependent NO release in bEND5 cells, an established model of brain endothelial cells. We found that glutamate induced a dose-dependent oscillatory increase in [Ca2+ ]i , which was maximally activated at 200 µM and inhibited by α-methyl-4-carboxyphenylglycine, a selective blocker of Group 1 metabotropic glutamate receptors. Glutamate-induced intracellular Ca2+ oscillations were triggered by rhythmic endogenous Ca2+ mobilization and maintained over time by extracellular Ca2+ entry. Pharmacological manipulation revealed that glutamate-induced endogenous Ca2+ release was mediated by InsP3 -sensitive receptors and nicotinic acid adenine dinucleotide phosphate (NAADP) gated two-pore channel 1. Constitutive store-operated Ca2+ entry mediated Ca2+ entry during ongoing Ca2+ oscillations. Finally, glutamate evoked a robust, although delayed increase in NO levels, which was blocked by pharmacologically inhibition of the accompanying intracellular Ca2+ signals. Of note, glutamate induced Ca2+ -dependent NO release also in hCMEC/D3 cells, an established model of human brain microvascular endothelial cells. This investigation demonstrates for the first time that metabotropic glutamate-induced intracellular Ca2+ oscillations and NO release have the potential to impact on neurovascular coupling in the brain.


Assuntos
Encéfalo/irrigação sanguínea , Sinalização do Cálcio/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Ácido Glutâmico/farmacologia , Inositol 1,4,5-Trifosfato/metabolismo , NADP/análogos & derivados , Acoplamento Neurovascular/efeitos dos fármacos , Óxido Nítrico/metabolismo , Animais , Canais de Cálcio/metabolismo , Linhagem Celular , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Humanos , Camundongos , NADP/metabolismo , Receptores de Glutamato Metabotrópico/agonistas , Fatores de Tempo
12.
J Cell Sci ; 130(16): 2736-2746, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28701513

RESUMO

The cellular prion protein (PrPC) whose conformational misfolding leads to the production of deadly prions, has a still-unclarified cellular function despite decades of intensive research. Following our recent finding that PrPC limits Ca2+ entry via store-operated Ca2+ channels in neurons, we investigated whether the protein could also control the activity of ionotropic glutamate receptors (iGluRs). To this end, we compared local Ca2+ movements in primary cerebellar granule neurons and cortical neurons transduced with genetically encoded Ca2+ probes and expressing, or not expressing, PrPC Our investigation demonstrated that PrPC downregulates Ca2+ entry through each specific agonist-stimulated iGluR and after stimulation by glutamate. We found that, although PrP-knockout (KO) mitochondria were displaced from the plasma membrane, glutamate addition resulted in a higher mitochondrial Ca2+ uptake in PrP-KO neurons than in their PrPC-expressing counterpart. This was because the increased Ca2+ entry through iGluRs in PrP-KO neurons led to a parallel increase in Ca2+-induced Ca2+ release via ryanodine receptor channels. These data thus suggest that PrPC takes part in the cell apparatus controlling Ca2+ homeostasis, and that PrPC is involved in protecting neurons from toxic Ca2+ overloads.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Ácido Glutâmico/farmacologia , Mitocôndrias/metabolismo , Neurônios/metabolismo , Proteínas Priônicas/fisiologia , Animais , Cálcio/toxicidade , Sinalização do Cálcio/genética , Células Cultivadas , Ácido Glutâmico/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neuroproteção/genética , Proteínas Priônicas/genética
13.
Eur J Neurosci ; 47(3): 211-221, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29283464

RESUMO

Astrocytes participate in the development and resolution of neuroinflammation in numerous ways, including the release of cytokines and growth factors. Among many, astrocytes release transforming growth factors beta (TGF-ß) TGF-ß1, TGF-ß2 and TGF-ß3. TGF-ß1 is the most studied isoform, while production and release of TGF-ß2 and TGF-ß3 by astrocytes have been poorly characterized. Here, we report that purified cultures of hippocampal astrocytes produce mainly TGF-ß3 followed by TGF-ß2 and TGF-ß1. Furthermore, astrocytes release principally the active form of TGF-ß3 over the other two. Changes in release of TGF-ß were sensitive to the calcineurin (CaN) inhibitor FK506. Starvation had no effect on TGF-ß1 and TGF-ß3 while TGF-ß2 mRNA was significantly up-regulated in a CaN-dependent manner. We further investigated production and release of astroglial TGF-ß in Alzheimer's disease-related conditions. Oligomeric ß-amyloid (Aß) down-regulated TGF-ß1, while up-regulating TGF-ß2 and TGF-ß3, in a CaN-dependent manner. In cultured hippocampal astrocytes from 3xTg-AD mice, TGF-ß2 and TGF-ß3, but not TGF-ß1, were up-regulated, and this was CaN-independent. In hippocampal tissues from symptomatic 3xTg-AD mice, TGF-ß2 was up-regulated with respect to control mice. Finally, treatment with recombinant TGF-ßs showed that TGF-ß2 and TGF-ß3 significantly reduced PSD95 protein in cultured hippocampal neurons, and this effect was paralleled by conditioned media from Aß-treated astrocytes or from astrocytes from 3xTg-AD mice. Taken together, our data suggest that TGF-ß2 and TGF-ß3 are produced by astrocytes in a CaN-dependent manner and should be investigated further in the context of astrocyte-mediated neurodegeneration.


Assuntos
Astrócitos/metabolismo , Neurônios/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Fator de Crescimento Transformador beta3/metabolismo , Doença de Alzheimer/metabolismo , Animais , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Camundongos , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo
14.
Int J Mol Sci ; 19(10)2018 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-30274399

RESUMO

Astrocytes respond to neuronal activity by generating calcium signals which are implicated in the regulation of astroglial housekeeping functions and/or in modulation of synaptic transmission. We hypothesized that activity-induced calcium signals in astrocytes may activate calcineurin (CaN), a calcium/calmodulin-regulated protein phosphatase, implicated in neuropathology, but whose role in astroglial physiology remains unclear. We used a lentiviral vector expressing NFAT-EYFP (NY) fluorescent calcineurin sensor and a chemical protocol of LTP induction (cLTP) to show that, in mixed neuron-astrocytic hippocampal cultures, cLTP induced robust NY translocation into astrocyte nuclei and, hence, CaN activation. NY translocation was abolished by the CaN inhibitor FK506, and was not observed in pure astroglial cultures. Using Fura-2 single cell calcium imaging, we found sustained Ca2+ elevations in juxtaneuronal, but not distal, astrocytes. Pharmacological analysis revealed that both the Ca2+ signals and the nuclear NY translocation in astrocytes required NMDA and mGluR5 receptors and depended on extracellular Ca2+ entry via a store-operated mechanism. Our results provide a proof of principle that calcineurin in astrocytes may be activated in response to neuronal activity, thereby delineating a framework for investigating the role of astroglial CaN in the physiology of central nervous system.


Assuntos
Astrócitos/metabolismo , Calcineurina/metabolismo , Hipocampo/citologia , Neuroglia/metabolismo , Neurônios/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Cálcio/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Citosol/metabolismo , Vetores Genéticos/metabolismo , Glicina/análogos & derivados , Glicina/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Modelos Biológicos , N-Metilaspartato/metabolismo , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptor de Glutamato Metabotrópico 5/metabolismo , Reprodutibilidade dos Testes , Resorcinóis/farmacologia
15.
J Cell Biochem ; 117(10): 2260-71, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26917354

RESUMO

Truly endothelial progenitor cells (EPCs) can be mobilized from bone marrow to support the vascular network of growing tumors, thereby sustaining the metastatic switch. Endothelial colony forming cells (ECFCs) are the only EPC subtype belonging to the endothelial phenotype and capable of incorporating within neovessels. The intracellular Ca(2+) machinery plays a key role in ECFC activation and is remodeled in renal cellular carcinoma-derived ECFCs (RCC-ECFCs). Particularly, RCC-ECFCs seems to undergo a drop in endoplasmic reticulum (ER) Ca(2+) concentration ([Ca(2+) ]ER ). This feature is remarkable when considering that inositol-1,4,5-trisphosphate (InsP3 )-dependent ER-to-mitochondria Ca(2+) transfer regulates the intrinsic apoptosis pathway. Herein, we sought to assess whether: (1) the [Ca(2+) ]ER and the InsP3 -induced ER-mitochondria Ca(2+) shuttle are reduced in RCC-ECFCs; and (2) the dysregulation of ER Ca(2+) handling leads to apoptosis resistance in tumor-derived cells. RCC-ECFCs displayed a reduction both in [Ca(2+) ]ER and in the InsP3 -dependent mitochondrial Ca(2+) uptake, while they expressed normal levels of Bcl-2 and Bak. The decrease in [Ca(2+) ]ER was associated to a remarkable ER expansion in RCC-ECFCs, which is a hallmark of ER stress, and did not depend on the remodeling of the Ca(2+) -transporting and the ER Ca(2+) -storing systems. As expected, RCC-ECFCs were less sensitive to rapamycin- and thapsigargin-induced apoptosis; however, buffering intracellular Ca(2+) levels with BAPTA dampened apoptosis in both cell types. Finally, store-operated Ca(2+) entry was seemingly uncoupled from the apoptotic machinery in RCC-ECFCs. Thus, the chronic underfilling of the ER Ca(2+) pool could confer a survival advantage to RCC-ECFCs and underpin RCC resistance to pharmacological treatment. J. Cell. Biochem. 117: 2260-2271, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Apoptose , Cálcio/metabolismo , Carcinoma de Células Renais/patologia , Retículo Endoplasmático/patologia , Células Progenitoras Endoteliais/patologia , Neoplasias Renais/patologia , Mitocôndrias/patologia , Adulto , Carcinoma de Células Renais/metabolismo , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas , Retículo Endoplasmático/metabolismo , Células Progenitoras Endoteliais/metabolismo , Feminino , Humanos , Neoplasias Renais/metabolismo , Masculino , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Transdução de Sinais , Adulto Jovem
16.
Artigo em Inglês | MEDLINE | ID: mdl-24935225

RESUMO

The most accredited (and fashionable) hypothesis of the pathogenesis of Alzheimer Disease (AD) sees accumulation of ß-amyloid protein in the brain (in both soluble and insoluble forms) as a leading mechanism of neurotoxicity. How ß-amyloid triggers the neurodegenerative disorder is at present unclear, but growing evidence suggests that a deregulation of Ca(2+) homeostasis and deficient Ca(2+) signalling may represent a fundamental pathogenic factor. Given that symptoms of AD are most likely linked to synaptic dysfunction (at the early stages) followed by neuronal loss (at later and terminal phases of the disease), the effects of ß-amyloid have been mainly studied in neurones. Yet, it must be acknowledged that neuroglial cells, including astrocytes, contribute to pathological progression of most (if not all) neurological diseases. Here, we review the literature pertaining to changes in Ca(2+) signalling in astrocytes exposed to exogenous ß-amyloid or in astrocytes from transgenic Alzheimer disease animals models, characterized by endogenous ß-amyloidosis. Accumulated experimental data indicate deregulation of Ca(2+) homeostasis and signalling in astrocytes in AD, which should be given full pathogenetic consideration. Further studies are warranted to comprehend the role of deficient astroglial Ca(2+) signalling in the disease progression.


Assuntos
Doença de Alzheimer/metabolismo , Sinalização do Cálcio , Neuroglia/metabolismo , Peptídeos beta-Amiloides/fisiologia , Animais , Humanos
18.
Glia ; 61(7): 1134-45, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23616440

RESUMO

The amyloid hypothesis of Alzheimer's disease (AD) suggests that soluble amyloid ß (Aß) is an initiator of a cascade of events eventually leading to neurodegeneration. Recently, we reported that Aß deranged Ca(2+) homeostasis specifically in hippocampal astrocytes by targeting key elements of Ca(2+) signaling, such as mGluR5 and IP3 R1. In the present study, we dissect a cascade of signaling events by which Aß deregulates glial Ca(2+) : (i) 100 nM Aß leads to an increase in cytosolic calcium after 4-6 h of treatment; (ii) mGluR5 is increased after 24 h of treatment; (iii) this increase is blocked by inhibitors of calcineurin (CaN) and NF-kB. Furthermore, we show that Aß treatment of glial cells leads to de-phosphorylation of Bcl10 and an increased CaN-Bcl10 interaction. Last, mGluR5 staining is augmented in hippocampal astrocytes of AD patients in proximity of Aß plaques and co-localizes with nuclear accumulation of the p65 NF-kB subunit and increased staining of CaNAα. Taken together our data suggest that nanomolar [Aß] deregulates Ca(2+) homeostasis via CaN and its downstream target NF-kB, possibly via the cross-talk of Bcl10 in hippocampal astrocytes.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Astrócitos/efeitos dos fármacos , Calcineurina/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , NF-kappa B/metabolismo , Fragmentos de Peptídeos/farmacologia , Receptor de Glutamato Metabotrópico 5/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Animais , Animais Recém-Nascidos , Proteína 10 de Linfoma CCL de Células B , Compostos de Boro/farmacologia , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Células Cultivadas , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Hipocampo/citologia , Hipocampo/metabolismo , Humanos , Masculino , Proteínas de Neoplasias/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Ratos , Receptor de Glutamato Metabotrópico 5/genética , Regulação para Cima/efeitos dos fármacos
19.
Eur J Clin Invest ; 43(10): 1039-51, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23937291

RESUMO

BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative disorder featured by deposition of beta-amyloid (Aß) plaques in the hippocampus and associated cortices and progressive cognitive decline. Tropisetron, a selective 5-HT3 receptor antagonist, is conventionally used to counteract chemotherapy-induced emesis. Recent investigations describe antiphlogistic properties for tropisetron. It has been shown that tropisetron protects against rat embolic stroke. We investigated protective properties of tropisetron in a beta-amyloid (Aß) rat model of AD and possible involvement of 5-HT3 receptors. MATERIAL AND METHODS: Aß (1-42) was injected into the hippocampus of male rats. Animals were treated intracerebroventricularly with tropisetron, mCPBG (selective 5-HT3 receptor agonist) or mCPBG plus tropisetron on days 1, 3, 5 and 7. Seven days following Aß administration, inflammatory markers (TNF-α, COX-2, iNOS and NF-κB), apoptotic markers (caspase 3 cytochrome c release) and calcineurin phosphatase activity were assessed in hippocampus. RESULTS: Seven days following Aß inoculation, control animals displayed dramatic increase in TNF-α, COX-2, iNOS, NF-κB, active caspase 3, cytochrome c release and calcineurin phosphatase activity in the hippocampus. Tropisetron significantly diminished the elevated levels of these markers and reversed the cognitive deficit. Interestingly, tropisetron was also found to be a potent inhibitor of calcineurin phosphatase activity. The selective 5-HT3 receptor agonist mCPBG, when co-administered with tropisetron, completely reversed the procognitive and anti-apoptotic properties of tropisetron while it could only partially counteract the anti-inflammatory effects. mCPBG alone significantly aggravated Aß-induced injury. CONCLUSION: Our findings indicate that tropisetron protects against Aß-induced neurotoxicity in vivo through both 5-HT3 receptor-dependent and independent pathways.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Apoptose/efeitos dos fármacos , Indóis/farmacologia , Antagonistas do Receptor 5-HT3 de Serotonina/farmacologia , Peptídeos beta-Amiloides/toxicidade , Animais , Calcineurina/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Citocromos c/metabolismo , Encefalite/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Nitritos/metabolismo , Ratos , Ratos Wistar , Tropizetrona , Fator de Necrose Tumoral alfa/metabolismo
20.
Neural Regen Res ; 18(8): 1672-1678, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36751778

RESUMO

Alzheimer's disease (AD) is characterized by complex etiology, long-lasting pathogenesis, and cell-type-specific alterations. Currently, there is no cure for AD, emphasizing the urgent need for a comprehensive understanding of cell-specific pathology. Astrocytes, principal homeostatic cells of the central nervous system, are key players in the pathogenesis of neurodegenerative diseases, including AD. Cellular models greatly facilitate the investigation of cell-specific pathological alterations and the dissection of molecular mechanisms and pathways. Tumor-derived and immortalized astrocytic cell lines, alongside the emerging technology of adult induced pluripotent stem cells, are widely used to study cellular dysfunction in AD. Surprisingly, no stable cell lines were available from genetic mouse AD models. Recently, we established immortalized hippocampal astroglial cell lines from amyloid-ß precursor protein/presenilin-1/Tau triple-transgenic (3xTg)-AD mice (denominated as wild type (WT)- and 3Tg-iAstro cells) using retrovirus-mediated transduction of simian virus 40 large T-antigen and propagation without clonal selection, thereby maintaining natural heterogeneity of primary cultures. Several groups have successfully used 3Tg-iAstro cells for single-cell and omics approaches to study astrocytic AD-related alterations of calcium signaling, mitochondrial dysfunctions, disproteostasis, altered homeostatic and signaling support to neurons, and blood-brain barrier models. Here we provide a comparative overview of the most used models to study astrocytes in vitro, such as primary culture, tumor-derived cell lines, immortalized astroglial cell lines, and induced pluripotent stem cell-derived astrocytes. We conclude that immortalized WT- and 3Tg-iAstro cells provide a non-competitive but complementary, low-cost, easy-to-handle, and versatile cellular model for dissection of astrocyte-specific AD-related alterations and preclinical drug discovery.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA