Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurochem ; 2023 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-37804203

RESUMO

Significant evidence suggests that misfolded alpha-synuclein (aSyn), a major component of Lewy bodies, propagates in a prion-like manner contributing to disease progression in Parkinson's disease (PD) and other synucleinopathies. In fact, timed inoculation of M83 hemizygous mice with recombinant human aSyn preformed fibrils (PFF) has shown symptomatic deficits after substantial spreading of pathogenic alpha-synuclein, as detected by markers for the phosphorylation of S129 of aSyn. However, whether accumulated toxicity impact human-relevant cognitive and structural neuroanatomical measures is not fully understood. Here we performed a single unilateral striatal PFF injection in M83 hemizygous mice, and using two assays with translational potential, ex vivo magnetic resonance imaging (MRI) and touchscreen testing, we examined the combined neuroanatomical and behavioral impact of aSyn propagation. In PFF-injected mice, we observed widespread atrophy in bilateral regions that project to or receive input from the injection site using MRI. We also identified early deficits in reversal learning prior to the emergence of motor symptoms. Our findings highlight a network of regions with related cellular correlates of pathology that follow the progression of aSyn spreading, and that affect brain areas relevant for reversal learning. Our experiments suggest that M83 hemizygous mice injected with human PFF provides a model to understand how misfolded aSyn affects human-relevant pre-clinical measures and suggest that these pre-clinical biomarkers could be used to detect early toxicity of aSyn and provide better translational measures between mice and human disease.

2.
Acta Neuropathol ; 144(5): 881-910, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36121476

RESUMO

The predominantly pre-synaptic intrinsically disordered protein α-synuclein is prone to misfolding and aggregation in synucleinopathies, such as Parkinson's disease (PD) and Dementia with Lewy bodies (DLB). Molecular chaperones play important roles in protein misfolding diseases and members of the chaperone machinery are often deposited in Lewy bodies. Here, we show that the Hsp90 co-chaperone STI1 co-immunoprecipitated α-synuclein, and co-deposited with Hsp90 and Hsp70 in insoluble protein fractions in two mouse models of α-synuclein misfolding. STI1 and Hsp90 also co-localized extensively with filamentous S129 phosphorylated α-synuclein in ubiquitin-positive inclusions. In PD human brains, STI1 transcripts were increased, and in neurologically healthy brains, STI1 and α-synuclein transcripts correlated. Nuclear Magnetic Resonance (NMR) analyses revealed direct interaction of α-synuclein with STI1 and indicated that the STI1 TPR2A, but not TPR1 or TPR2B domains, interacted with the C-terminal domain of α-synuclein. In vitro, the STI1 TPR2A domain facilitated S129 phosphorylation by Polo-like kinase 3. Moreover, mice over-expressing STI1 and Hsp90ß presented elevated α-synuclein S129 phosphorylation accompanied by inclusions when injected with α-synuclein pre-formed fibrils. In contrast, reduced STI1 function decreased protein inclusion formation, S129 α-synuclein phosphorylation, while mitigating motor and cognitive deficits as well as mesoscopic brain atrophy in α-synuclein-over-expressing mice. Our findings reveal a vicious cycle in which STI1 facilitates the generation and accumulation of toxic α-synuclein conformers, while α-synuclein-induced proteostatic stress increased insoluble STI1 and Hsp90.


Assuntos
Proteínas de Choque Térmico/metabolismo , Proteínas Intrinsicamente Desordenadas , alfa-Sinucleína/metabolismo , Animais , Proteínas de Choque Térmico HSP90/química , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Camundongos , Chaperonas Moleculares/metabolismo , Fosfoproteínas , Ubiquitinas , alfa-Sinucleína/toxicidade
3.
eNeuro ; 9(5)2022.
Artigo em Inglês | MEDLINE | ID: mdl-36635920

RESUMO

The protease caspase-3 is a key mediator of apoptotic programmed cell death. But weak or transient caspase activity can contribute to neuronal differentiation, axonal pathfinding, and synaptic long-term depression. Despite the importance of sublethal, or nonapoptotic, caspase activity in neurodevelopment and neural plasticity, there has been no simple method for mapping and quantifying nonapoptotic caspase activity (NACA) in rodent brains. We therefore generated a transgenic mouse expressing a highly sensitive and specific fluorescent reporter of caspase activity, with peak signal localized to the nucleus. As a proof of concept, we first obtained evidence that NACA influences neurophysiology in an amygdalar circuit. Then focusing on the amygdala, we were able to quantify a sex-specific persistent elevation in caspase activity in females after restraint stress. This simple in vivo caspase activity reporter will facilitate systems-level studies of apoptotic and nonapoptotic phenomena in behavioral and pathologic models.


Assuntos
Apoptose , Encéfalo , Masculino , Feminino , Camundongos , Animais , Apoptose/fisiologia , Camundongos Transgênicos , Plasticidade Neuronal , Caspase 9
4.
FASEB J ; 35(5): e21594, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33908654

RESUMO

Protein misfolding is a central feature of most neurodegenerative diseases. Molecular chaperones can modulate the toxicity associated with protein misfolding, but it remains elusive which molecular chaperones and co-chaperones interact with specific misfolded proteins. TDP-43 misfolding and inclusion formation are a hallmark of amyotrophic lateral sclerosis (ALS) and other neurodegenerative diseases. Using yeast and mammalian neuronal cells we find that Hsp90 and its co-chaperone Sti1 have the capacity to alter TDP-43 misfolding, inclusion formation, aggregation, and cellular toxicity. Our data also demonstrate that impaired Hsp90 function sensitizes cells to TDP-43 toxicity and that Sti1 specifically interacts with and strongly modulates TDP-43 toxicity in a dose-dependent manner. Our study thus uncovers a previously unrecognized tie between Hsp90, Sti1, TDP-43 misfolding, and cellular toxicity.


Assuntos
Apoptose , Proteínas de Ligação a DNA/química , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico/fisiologia , Dobramento de Proteína , Saccharomyces cerevisiae/metabolismo , Proteinopatias TDP-43/patologia , Animais , Sítios de Ligação , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Proteínas de Choque Térmico HSP90/genética , Células HeLa , Humanos , Corpos de Inclusão , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteinopatias TDP-43/etiologia
5.
J Oral Pathol Med ; 50(10): 985-994, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33896033

RESUMO

BACKGROUND: Head and neck squamous cell carcinoma (HNSC) etiopathogenesis remains unclear, and the biological changes with the activation of heat shock proteins (HSPs) and prion protein (PRNP) promoted by hypoxia in HNSC are undetermined. This study investigates hypoxia's effect in lymph node metastasis by PRNP expression changes and its main partners. METHODS: The study combined a theoretical/cell culture study with a case-control study. First, bioinformatics and cell culture were performed. A case-control study was performed in a second step by comparing HNSC patients with and without lymph node metastasis. ANALYSES: The Cancer Genome Atlas (TCGA) data source validates the theory in the global population study. RESULTS: Bioinformatics analysis suggests that hypoxia-inducible factor-1α (HIF1A) is associated with HSPA4, HSP90AA1 and PRNP expression. TCGA data validate the hypothesis that higher HSP90AA1, HSPA4 and PRNP are related to metastases and low survival. Herein, the cell study demonstrated that muted PRNP did not respond to hypoxia. DISCUSSION: Our results collectively provide the first evidence that PRNP promotes HNSC lymph node metastasis progression through the upregulation of HSPA4, HSP90AA1 and HIF1A. Our findings may provide a molecular basis for the promoting Role of PRNP in HNSC progression.


Assuntos
Neoplasias de Cabeça e Pescoço , Proteínas Priônicas , Biomarcadores Tumorais/genética , Estudos de Casos e Controles , Neoplasias de Cabeça e Pescoço/genética , Humanos , Proteínas Priônicas/genética , Prognóstico , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética
6.
Acta Neuropathol Commun ; 9(1): 60, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33823944

RESUMO

We have previously reported long-term changes in the brains of non-concussed varsity rugby players using magnetic resonance spectroscopy (MRS), diffusion tensor imaging (DTI) and functional magnetic imaging (fMRI). Others have reported cognitive deficits in contact sport athletes that have not met the diagnostic criteria for concussion. These results suggest that repetitive mild traumatic brain injuries (rmTBIs) that are not severe enough to meet the diagnostic threshold for concussion, produce long-term consequences. We sought to characterize the neuroimaging, cognitive, pathological and metabolomic changes in a mouse model of rmTBI. Using a closed-skull model of mTBI that when scaled to human leads to rotational and linear accelerations far below what has been reported for sports concussion athletes, we found that 5 daily mTBIs triggered two temporally distinct types of pathological changes. First, during the first days and weeks after injury, the rmTBI produced diffuse axonal injury, a transient inflammatory response and changes in diffusion tensor imaging (DTI) that resolved with time. Second, the rmTBI led to pathological changes that were evident months after the injury including: changes in magnetic resonance spectroscopy (MRS), altered levels of synaptic proteins, behavioural deficits in attention and spatial memory, accumulations of pathologically phosphorylated tau, altered blood metabolomic profiles and white matter ultrastructural abnormalities. These results indicate that exceedingly mild rmTBI, in mice, triggers processes with pathological consequences observable months after the initial injury.


Assuntos
Concussão Encefálica/patologia , Concussão Encefálica/fisiopatologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Animais , Comportamento Animal , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Genes Brain Behav ; 20(1): e12650, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32141694

RESUMO

The rise in the number of users and institutions utilizing the rodent touchscreen technology for cognitive testing over the past decade has prompted the need for knowledge mobilization and community building. To address the needs of the growing touchscreen community, the first international touchscreen symposium was hosted at Western University. Attendees from around the world attended talks from expert neuroscientists using touchscreens to examine a vast array of questions regarding cognition and the nervous system. In addition to the symposium, a subset of attendees was invited to partake in a hands-on training course where they received touchscreen training covering both hardware and software components. Beyond the two touchscreen events, virtual platforms have been developed to further support touchscreen users: (a) Mousebytes.ca, which includes a data repository of rodent touchscreen tasks, and (b) Touchscreencognition.org, an online community with numerous training and community resources, perhaps most notably a forum where members can ask and answer questions. The advantages of the rodent touchscreen technology for cognitive neuroscience research has allowed neuroscientists from diverse backgrounds to test specific cognitive processes using well-validated and standardized apparatus, contributing to its rise in popularity and its relevance to modern neuroscience research. The commitment of the touchscreen community to data, task development and information sharing not only ensures an expansive future of the use of rodent touchscreen technology but additionally, quality research that will increase translation from preclinical studies to clinical successes.


Assuntos
Pesquisa Comportamental/métodos , Cognição , Roedores/fisiologia , Interface Usuário-Computador , Animais , Pesquisa Comportamental/instrumentação , Pesquisa Comportamental/estatística & dados numéricos , Congressos como Assunto , Roedores/genética , Roedores/psicologia , Tato
8.
J Neurochem ; 153(6): 727-758, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31562773

RESUMO

Chaperone networks are dysregulated with aging, but whether compromised Hsp70/Hsp90 chaperone function disturbs neuronal resilience is unknown. Stress-inducible phosphoprotein 1 (STI1; STIP1; HOP) is a co-chaperone that simultaneously interacts with Hsp70 and Hsp90, but whose function in vivo remains poorly understood. We combined in-depth analysis of chaperone genes in human datasets, analysis of a neuronal cell line lacking STI1 and of a mouse line with a hypomorphic Stip1 allele to investigate the requirement for STI1 in aging. Our experiments revealed that dysfunctional STI1 activity compromised Hsp70/Hsp90 chaperone network and neuronal resilience. The levels of a set of Hsp90 co-chaperones and client proteins were selectively affected by reduced levels of STI1, suggesting that their stability depends on functional Hsp70/Hsp90 machinery. Analysis of human databases revealed a subset of co-chaperones, including STI1, whose loss of function is incompatible with life in mammals, albeit they are not essential in yeast. Importantly, mice expressing a hypomorphic STI1 allele presented spontaneous age-dependent hippocampal neurodegeneration and reduced hippocampal volume, with consequent spatial memory deficit. We suggest that impaired STI1 function compromises Hsp70/Hsp90 chaperone activity in mammals and can by itself cause age-dependent hippocampal neurodegeneration in mice. Cover Image for this issue: doi: 10.1111/jnc.14749.


Assuntos
Envelhecimento/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico/deficiência , Hipocampo/metabolismo , Chaperonas Moleculares/metabolismo , Adaptação Fisiológica/fisiologia , Envelhecimento/genética , Animais , Células-Tronco Embrionárias/metabolismo , Técnicas de Inativação de Genes/métodos , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico/genética , Hipocampo/citologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Chaperonas Moleculares/genética , Neurônios/metabolismo
9.
Elife ; 82019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31825307

RESUMO

Open Science has changed research by making data accessible and shareable, contributing to replicability to accelerate and disseminate knowledge. However, for rodent cognitive studies the availability of tools to share and disseminate data is scarce. Automated touchscreen-based tests enable systematic cognitive assessment with easily standardised outputs that can facilitate data dissemination. Here we present an integration of touchscreen cognitive testing with an open-access database public repository (mousebytes.ca), as well as a Web platform for knowledge dissemination (https://touchscreencognition.org). We complement these resources with the largest dataset of age-dependent high-level cognitive assessment of mouse models of Alzheimer's disease, expanding knowledge of affected cognitive domains from male and female mice of three strains. We envision that these new platforms will enhance sharing of protocols, data availability and transparency, allowing meta-analysis and reuse of mouse cognitive data to increase the replicability/reproducibility of datasets.


Assuntos
Cognição/fisiologia , Ciência dos Animais de Laboratório/instrumentação , Ciência dos Animais de Laboratório/métodos , Roedores , Doença de Alzheimer , Animais , Comportamento Animal , Comportamento de Escolha , Bases de Dados Factuais , Modelos Animais de Doenças , Feminino , Aprendizagem/fisiologia , Masculino , Memória/fisiologia , Camundongos , Testes Neuropsicológicos , Reprodutibilidade dos Testes , Roedores/genética , Software
10.
J Neurochem ; 145(1): 68-79, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29265373

RESUMO

Stress-inducible phosphoprotein 1 (STI1) acts as a neuroprotective factor in the ischemic brain and its levels are increased following ischemia. Previous work has suggested that some of these STI1 actions in a stroke model depend on the recruitment of bone marrow-derived stem cells to improve outcomes after ischemic insult. However, STI1 can directly increase neuroprotective signaling in neurons by engaging with the cellular prion protein (PrPC ) and activating α7 nicotinic acetylcholine receptors (α7nAChR). Given that α7nAChR activation has also been involved in neuroprotection in stroke, it is possible that STI1 can have direct actions on neurons to prevent deleterious consequences of ischemic insults. Here, we tested this hypothesis by exposing primary neuronal cultures to 1-h oxygen-glucose deprivation (OGD) and reperfusion and assessing signaling pathways activated by STI1/PrPC . Our results demonstrated that STI1 treatment significantly decreased apoptosis and cell death in mouse neurons submitted to OGD in a manner that was dependent on PrPC and α7nAChR, but also on the activin A receptor 1 (ALK2), which has emerged as a signaling partner of STI1. Interestingly, pharmacological inhibition of the ALK2 receptor prevented neuroprotection by STI1, while activation of ALK2 receptors by bone morphogenetic protein 4 (BMP4) either before or after OGD was effective in decreasing neuronal death induced by ischemia. We conclude that PrPC /STI1 engagement and its subsequent downstream signaling cascades involving α7nAChR as well as the ALK2 receptor may be activated in neurons by increased levels of STI1. This signaling pathway protects neurons from ischemic insults.


Assuntos
Isquemia Encefálica/metabolismo , Proteínas de Choque Térmico/metabolismo , Neuroproteção/fisiologia , Proteínas Priônicas/metabolismo , Receptores de Ativinas Tipo I/metabolismo , Animais , Apoptose/fisiologia , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Transdução de Sinais/fisiologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
11.
Biochem J ; 474(17): 2981-2991, 2017 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-28739602

RESUMO

Prion protein (PrPC) was initially described due to its involvement in transmissible spongiform encephalopathies. It was subsequently demonstrated to be a cell surface molecule involved in many physiological processes, such as vesicle trafficking. Here, we investigated the roles of PrPC in the response to insulin and obesity development. Two independent PrPC knockout (KO) and one PrPC overexpressing (TG20) mouse models were fed high-fat diets, and the development of insulin resistance and obesity was monitored. PrPC KO mice fed high-fat diets presented all of the symptoms associated with the development of insulin resistance: hyperglycemia, hyperinsulinemia, and obesity. Conversely, TG20 animals fed high-fat diets showed reduced weight and insulin resistance. Accordingly, the expression of peroxisome proliferator-activated receptor gamma (PPARγ) was reduced in PrPC KO mice and increased in TG20 animals. PrPC KO cells also presented reduced glucose uptake upon insulin stimulation, due to reduced translocation of the glucose transporter Glut4. Thus, our results suggest that PrPC reflects susceptibility to the development of insulin resistance and metabolic syndrome.


Assuntos
Transportador de Glucose Tipo 4/metabolismo , Resistência à Insulina , Obesidade/metabolismo , PPAR gama/metabolismo , Proteínas PrPC/metabolismo , Proteínas Priônicas/metabolismo , Células 3T3-L1 , Animais , Membrana Celular/metabolismo , Membrana Celular/patologia , Células Cultivadas , Cruzamentos Genéticos , Dieta Hiperlipídica/efeitos adversos , Embrião de Mamíferos/patologia , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Obesidade/etiologia , Obesidade/patologia , PPAR gama/genética , Proteínas PrPC/genética , Proteínas Priônicas/genética , Transporte Proteico , Aumento de Peso
12.
J Biol Chem ; 291(42): 21945-21955, 2016 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-27563063

RESUMO

The prion protein (PrPC) has been suggested to operate as a scaffold/receptor protein in neurons, participating in both physiological and pathological associated events. PrPC, laminin, and metabotropic glutamate receptor 5 (mGluR5) form a protein complex on the plasma membrane that can trigger signaling pathways involved in neuronal differentiation. PrPC and mGluR5 are co-receptors also for ß-amyloid oligomers (AßOs) and have been shown to modulate toxicity and neuronal death in Alzheimer's disease. In the present work, we addressed the potential crosstalk between these two signaling pathways, laminin-PrPC-mGluR5 or AßO-PrPC-mGluR5, as well as their interplay. Herein, we demonstrated that an existing complex containing PrPC-mGluR5 has an important role in AßO binding and activity in neurons. A peptide mimicking the binding site of laminin onto PrPC (Ln-γ1) binds to PrPC and induces intracellular Ca2+ increase in neurons via the complex PrPC-mGluR5. Ln-γ1 promotes internalization of PrPC and mGluR5 and transiently decreases AßO biding to neurons; however, the peptide does not impact AßO toxicity. Given that mGluR5 is critical for toxic signaling by AßOs and in prion diseases, we tested whether mGlur5 knock-out mice would be susceptible to prion infection. Our results show mild, but significant, effects on disease progression, without affecting survival of mice after infection. These results suggest that PrPC-mGluR5 form a functional response unit by which multiple ligands can trigger signaling. We propose that trafficking of PrPC-mGluR5 may modulate signaling intensity by different PrPC ligands.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas PrPC/metabolismo , Doenças Priônicas/metabolismo , Multimerização Proteica , Receptor de Glutamato Metabotrópico 5/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Cálcio/metabolismo , Sinalização do Cálcio/genética , Camundongos , Camundongos Knockout , Neurônios/patologia , Fragmentos de Peptídeos/genética , Proteínas PrPC/genética , Doenças Priônicas/genética , Doenças Priônicas/patologia , Transporte Proteico/genética , Receptor de Glutamato Metabotrópico 5/genética
13.
Biochem J ; 473(14): 2119-30, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27208175

RESUMO

Soluble oligomers of amyloid-beta peptide (AßO) transmit neurotoxic signals through the cellular prion protein (PrP(C)) in Alzheimer's disease (AD). Secreted stress-inducible phosphoprotein 1 (STIP1), an Hsp70 and Hsp90 cochaperone, inhibits AßO binding to PrP(C) and protects neurons from AßO-induced cell death. Here, we investigated the molecular interactions between AßO and STIP1 binding to PrP(C) and their effect on neuronal cell death. We showed that residues located in a short region of PrP (90-110) mediate AßO binding and we narrowed the major interaction in this site to amino acids 91-100. In contrast, multiple binding sites on STIP1 (DP1, TPR1 and TPR2A) contribute to PrP binding. DP1 bound the N-terminal of PrP (residues 23-95), whereas TPR1 and TPR2A showed binding to the C-terminal of PrP (residues 90-231). Importantly, only TPR1 and TPR2A directly inhibit both AßO binding to PrP and cell death. Furthermore, our structural studies reveal that TPR1 and TPR2A bind to PrP through distinct regions. The TPR2A interface was shown to be much more extensive and to partially overlap with the Hsp90 binding site. Our data show the possibility of a PrP, STIP1 and Hsp90 ternary complex, which may influence AßO-mediated cell death.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Proteínas PrPC/metabolismo , Animais , Sítios de Ligação , Morte Celular/genética , Morte Celular/fisiologia , Células Cultivadas , Proteínas de Choque Térmico/genética , Hipocampo/citologia , Espectroscopia de Ressonância Magnética , Camundongos , Neurônios/metabolismo , Ligação Proteica , Domínios Proteicos/genética , Domínios Proteicos/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
14.
J Neurosci ; 35(45): 15157-69, 2015 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-26558786

RESUMO

In Alzheimer's disease, accumulation of soluble oligomers of ß-amyloid peptide is known to be highly toxic, causing disturbances in synaptic activity and neuronal death. Multiple studies relate these effects to increased oxidative stress and aberrant activity of calcium-permeable cation channels leading to calcium imbalance. The transient receptor potential melastatin 2 (TRPM2) channel, a Ca(2+)-permeable nonselective cation channel activated by oxidative stress, has been implicated in neurodegenerative diseases, and more recently in amyloid-induced toxicity. Here we show that the function of TRPM2 is augmented by treatment of cultured neurons with ß-amyloid oligomers. Aged APP/PS1 Alzheimer's mouse model showed increased levels of endoplasmic reticulum stress markers, protein disulfide isomerase and phosphorylated eukaryotic initiation factor 2α, as well as decreased levels of the presynaptic marker synaptophysin. Elimination of TRPM2 in APP/PS1 mice corrected these abnormal responses without affecting plaque burden. These effects of TRPM2 seem to be selective for ß-amyloid toxicity, as ER stress responses to thapsigargin or tunicamycin in TRPM2(-/-) neurons was identical to that of wild-type neurons. Moreover, reduced microglial activation was observed in TRPM2(-/-)/APP/PS1 hippocampus compared with APP/PS1 mice. In addition, age-dependent spatial memory deficits in APP/PS1 mice were reversed in TRPM2(-/-)/APP/PS1 mice. These results reveal the importance of TRPM2 for ß-amyloid neuronal toxicity, suggesting that TRPM2 activity could be potentially targeted to improve outcomes in Alzheimer's disease. SIGNIFICANCE STATEMENT: Transient receptor potential melastatin 2 (TRPM2) is an oxidative stress sensing calcium-permeable channel that is thought to contribute to calcium dysregulation associated with neurodegenerative diseases, including Alzheimer's disease. Here we show that oligomeric ß-amyloid, the toxic peptide in Alzheimer's disease, facilitates TRPM2 channel activation. In mice designed to model Alzheimer's disease, genetic elimination of TRPM2 normalized deficits in synaptic markers in aged mice. Moreover, the absence of TRPM2 improved age-dependent spatial memory deficits observed in Alzheimer's mice. Our results reveal the importance of TRPM2 for neuronal toxicity and memory impairments in an Alzheimer's mouse model and suggest that TRPM2 could be targeted for the development of therapeutic agents effective in the treatment of dementia.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Transtornos da Memória/metabolismo , Canais de Cátion TRPM/deficiência , Doença de Alzheimer/induzido quimicamente , Peptídeos beta-Amiloides/toxicidade , Animais , Células Cultivadas , Humanos , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Placa Amiloide/metabolismo , Canais de Cátion TRPM/genética
15.
Dis Model Mech ; 8(11): 1457-66, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26398952

RESUMO

Stress-inducible phosphoprotein I (STIP1, STI1 or HOP) is a co-chaperone intermediating Hsp70/Hsp90 exchange of client proteins, but it can also be secreted to trigger prion protein-mediated neuronal signaling. Some mothers of children with autism spectrum disorders (ASD) present antibodies against certain brain proteins, including antibodies against STIP1. Maternal antibodies can cross the fetus blood-brain barrier during pregnancy, suggesting the possibility that they can interfere with STIP1 levels and, presumably, functions. However, it is currently unknown whether abnormal levels of STIP1 have any impact in ASD-related behavior. Here, we used mice with reduced (50%) or increased STIP1 levels (fivefold) to test for potential ASD-like phenotypes. We found that increased STIP1 regulates the abundance of Hsp70 and Hsp90, whereas reduced STIP1 does not affect Hsp70, Hsp90 or the prion protein. Interestingly, BAC transgenic mice presenting fivefold more STIP1 show no major phenotype when examined in a series of behavioral tasks, including locomotor activity, elevated plus maze, Morris water maze and five-choice serial reaction time task (5-CSRTT). In contrast, mice with reduced STIP1 levels are hyperactive and have attentional deficits on the 5-CSRTT, but exhibit normal performance for the other tasks. We conclude that reduced STIP1 levels can contribute to phenotypes related to ASD. However, future experiments are needed to define whether it is decreased chaperone capacity or impaired prion protein signaling that contributes to these phenotypes.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade/metabolismo , Comportamento Animal , Proteínas de Choque Térmico/deficiência , Animais , Transtorno do Deficit de Atenção com Hiperatividade/genética , Transtorno do Deficit de Atenção com Hiperatividade/psicologia , Modelos Animais de Doenças , Predisposição Genética para Doença , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico/genética , Masculino , Aprendizagem em Labirinto , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora , Fenótipo , Proteínas PrPC/metabolismo , Tempo de Reação , Natação , Fatores de Tempo
16.
J Neurosci ; 33(42): 16552-64, 2013 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-24133259

RESUMO

In Alzheimer's disease (AD), soluble amyloid-ß oligomers (AßOs) trigger neurotoxic signaling, at least partially, via the cellular prion protein (PrP(C)). However, it is unknown whether other ligands of PrP(C) can regulate this potentially toxic interaction. Stress-inducible phosphoprotein 1 (STI1), an Hsp90 cochaperone secreted by astrocytes, binds to PrP(C) in the vicinity of the AßO binding site to protect neurons against toxic stimuli. Here, we investigated a potential role of STI1 in AßO toxicity. We confirmed the specific binding of AßOs and STI1 to the PrP and showed that STI1 efficiently inhibited AßO binding to PrP in vitro (IC50 of ∼70 nm) and also decreased AßO binding to cultured mouse primary hippocampal neurons. Treatment with STI1 prevented AßO-induced synaptic loss and neuronal death in mouse cultured neurons and long-term potentiation inhibition in mouse hippocampal slices. Interestingly, STI1-haploinsufficient neurons were more sensitive to AßO-induced cell death and could be rescued by treatment with recombinant STI1. Noteworthy, both AßO binding to PrP(C) and PrP(C)-dependent AßO toxicity were inhibited by TPR2A, the PrP(C)-interacting domain of STI1. Additionally, PrP(C)-STI1 engagement activated α7 nicotinic acetylcholine receptors, which participated in neuroprotection against AßO-induced toxicity. We found an age-dependent upregulation of cortical STI1 in the APPswe/PS1dE9 mouse model of AD and in the brains of AD-affected individuals, suggesting a compensatory response. Our findings reveal a previously unrecognized role of the PrP(C) ligand STI1 in protecting neurons in AD and suggest a novel pathway that may help to offset AßO-induced toxicity.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Proteínas de Choque Térmico/metabolismo , Neurônios/metabolismo , Proteínas PrPC/metabolismo , Doença de Alzheimer/metabolismo , Animais , Astrócitos/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Hipocampo/metabolismo , Camundongos , Ligação Proteica , Transdução de Sinais/fisiologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
17.
Mol Cell Proteomics ; 12(11): 3253-70, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23938469

RESUMO

Stress-inducible phosphoprotein 1 (STI1), a cochaperone for Hsp90, has been shown to regulate multiple pathways in astrocytes, but its contributions to cellular stress responses are not fully understood. We show that in response to irradiation-mediated DNA damage stress STI1 accumulates in the nucleus of astrocytes. Also, STI1 haploinsufficiency decreases astrocyte survival after irradiation. Using yeast two-hybrid screenings we identified several nuclear proteins as STI1 interactors. Overexpression of one of these interactors, PIAS1, seems to be specifically involved in STI1 nuclear retention and in directing STI1 and Hsp90 to specific sub-nuclear regions. PIAS1 and STI1 co-immunoprecipitate and PIAS1 can function as an E3 SUMO ligase for STI. Using mass spectrometry we identified five SUMOylation sites in STI1. A STI1 mutant lacking these five sites is not SUMOylated, but still accumulates in the nucleus in response to increased expression of PIAS1, suggesting the possibility that a direct interaction with PIAS1 could be responsible for STI1 nuclear retention. To test this possibility, we mapped the interaction sites between PIAS1 and STI1 using yeast-two hybrid assays and surface plasmon resonance and found that a large domain in the N-terminal region of STI1 interacts with high affinity with amino acids 450-480 of PIAS1. Knockdown of PIAS1 in astrocytes impairs the accumulation of nuclear STI1 in response to irradiation. Moreover, a PIAS1 mutant lacking the STI1 binding site is unable to increase STI1 nuclear retention. Interestingly, in human glioblastoma multiforme PIAS1 expression is increased and we found a significant correlation between increased PIAS1 expression and STI1 nuclear localization. These experiments provide evidence that direct interaction between STI1 and PIAS1 is involved in the accumulation of nuclear STI1. This retention mechanism could facilitate nuclear chaperone activity.


Assuntos
Astrócitos/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Animais , Astrócitos/citologia , Astrócitos/efeitos da radiação , Morte Celular/genética , Morte Celular/fisiologia , Morte Celular/efeitos da radiação , Núcleo Celular/metabolismo , Células Cultivadas , Dano ao DNA , Raios gama , Técnicas de Silenciamento de Genes , Células HEK293 , Haploinsuficiência , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Inibidoras de STAT Ativados/antagonistas & inibidores , Proteínas Inibidoras de STAT Ativados/genética , Mapas de Interação de Proteínas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Estresse Fisiológico , Sumoilação , Técnicas do Sistema de Duplo-Híbrido
18.
FASEB J ; 27(9): 3594-607, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23729591

RESUMO

Stress-inducible phosphoprotein 1 (STI1) is part of the chaperone machinery, but it also functions as an extracellular ligand for the prion protein. However, the physiological relevance of these STI1 activities in vivo is unknown. Here, we show that in the absence of embryonic STI1, several Hsp90 client proteins are decreased by 50%, although Hsp90 levels are unaffected. Mutant STI1 mice showed increased caspase-3 activation and 50% impairment in cellular proliferation. Moreover, placental disruption and lack of cellular viability were linked to embryonic death by E10.5 in STI1-mutant mice. Rescue of embryonic lethality in these mutants, by transgenic expression of the STI1 gene, supported a unique role for STI1 during embryonic development. The response of STI1 haploinsufficient mice to cellular stress seemed compromised, and mutant mice showed increased vulnerability to ischemic insult. At the cellular level, ischemia increased the secretion of STI1 from wild-type astrocytes by 3-fold, whereas STI1 haploinsufficient mice secreted half as much STI1. Interesting, extracellular STI1 prevented ischemia-mediated neuronal death in a prion protein-dependent way. Our study reveals essential roles for intracellular and extracellular STI1 in cellular resilience.


Assuntos
Embrião de Mamíferos/metabolismo , Proteínas de Choque Térmico/metabolismo , Isquemia/metabolismo , Chaperonas Moleculares/metabolismo , Príons/metabolismo , Animais , Blastocisto/metabolismo , Western Blotting , Fator de Transcrição CDX2 , Células Cultivadas , Feminino , Proteínas de Choque Térmico/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Técnicas In Vitro , Isquemia/genética , Camundongos , Camundongos Mutantes , Chaperonas Moleculares/genética , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Gravidez , Príons/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
19.
J Neurochem ; 127(3): 415-25, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23651058

RESUMO

Prion protein (PrP(C) ), a glycosylphosphatidylinositol-anchored protein corrupted in prion diseases, has been shown recently to interact with group I metabotropic glutamate receptors (mGluRs). Moreover, both PrP(C) and mGluRs were proposed to function as putative receptors for ß-amyloid in Alzheimer's disease. PrP(C) can be processed in neurons via α or ß-cleavage to produce PrP(C) fragments that are neuroprotective or toxic, respectively. We found PrP(C) α-cleavage to be 2-3 times higher in the cortex of APPswe/PS1dE9 mice, a mouse model of Alzheimer's disease. A similar age-dependent increase was observed for PrP(C) ß-cleavage. Moreover, we observed considerable age-dependent increase in cortical expression of mGluR1, but not mGluR5. Exposure of cortical neuronal cultures to ß-amyloid oligomers upregulated mGluR1 and PrP(C) α-cleavage, while activation of group I mGluRs increased PrP(C) shedding from the membrane, likely due to increased levels of a disintegrin and metalloprotease10, a key disintegrin for PrP(C) shedding. Interestingly, a similar increase in a disintegrin and metalloprotease10 was detected in the cortex of 9-month-old APPswe/PS1dE9 animals. Our experiments reveal novel and complex processing of PrP(C) in connection with mGluR overexpression that seems to be triggered by ß-amyloid peptides. Prion protein (PrP(C) ) and metabotropic glutamate receptors (mGluR) are implicated in Alzheimer's disease (AD). We found age-dependent increase in PrP(C) processing, ADAM10 and mGluR1 levels in AD mouse model. These changes could be reproduced in cultured cortical neurons treated with Aß peptide. Our findings suggest that increased levels of Aß can trigger compensatory responses that may affect neuronal toxicity.


Assuntos
Anti-Inflamatórios , Ácidos Docosa-Hexaenoicos/farmacologia , Inflamação/patologia , Inflamação/prevenção & controle , Fármacos Neuroprotetores , Animais
20.
Cell Mol Life Sci ; 70(17): 3211-27, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23543276

RESUMO

The co-chaperone stress-inducible protein 1 (STI1) is released by astrocytes, and has important neurotrophic properties upon binding to prion protein (PrP(C)). However, STI1 lacks a signal peptide and pharmacological approaches pointed that it does not follow a classical secretion mechanism. Ultracentrifugation, size exclusion chromatography, electron microscopy, vesicle labeling, and particle tracking analysis were used to identify three major types of extracellular vesicles (EVs) released from astrocytes with sizes ranging from 20-50, 100-200, and 300-400 nm. These EVs carry STI1 and present many exosomal markers, even though only a subpopulation had the typical exosomal morphology. The only protein, from those evaluated here, present exclusively in vesicles that have exosomal morphology was PrP(C). STI1 partially co-localized with Rab5 and Rab7 in endosomal compartments, and a dominant-negative for vacuolar protein sorting 4A (VPS4A), required for formation of multivesicular bodies (MVBs), impaired EV and STI1 release. Flow cytometry and PK digestion demonstrated that STI1 localized to the outer leaflet of EVs, and its association with EVs greatly increased STI1 activity upon PrP(C)-dependent neuronal signaling. These results indicate that astrocytes secrete a diverse population of EVs derived from MVBs that contain STI1 and suggest that the interaction between EVs and neuronal surface components enhances STI1-PrP(C) signaling.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Choque Térmico/metabolismo , Vesículas Secretórias/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Hipocampo/citologia , Immunoblotting , Camundongos , Proteínas PrPC/metabolismo , Vesículas Secretórias/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...