Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 11: 1226604, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37645251

RESUMO

Astrocytes play a critical role in the maintenance of a healthy central nervous system and astrocyte dysfunction has been implicated in various neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). There is compelling evidence that mouse and human ALS and ALS/FTD astrocytes can reduce the number of healthy wild-type motoneurons (MNs) in co-cultures or after treatment with astrocyte conditioned media (ACM), independently of their genotype. A growing number of studies have shown that soluble toxic factor(s) in the ACM cause non-cell autonomous MN death, including our recent identification of inorganic polyphosphate (polyP) that is excessively released from mouse primary astrocytes (SOD1, TARDBP, and C9ORF72) and human induced pluripotent stem cells (iPSC)-derived astrocytes (TARDBP) to kill MNs. However, others have reported that astrocytes carrying mutant TDP43 do not produce detectable MN toxicity. This controversy is likely to arise from the findings that human iPSC-derived astrocytes exhibit a rather immature and/or reactive phenotype in a number of studies. Here, we have succeeded in generating a highly homogenous population of functional quiescent mature astrocytes from control subject iPSCs. Using identical conditions, we also generated mature astrocytes from an ALS/FTD patient carrying the TDP43A90V mutation. These mutant TDP43 patient-derived astrocytes exhibit key pathological hallmarks, including enhanced cytoplasmic TDP-43 and polyP levels. Additionally, mutant TDP43 astrocytes displayed a mild reactive signature and an aberrant function as they were unable to promote synaptogenesis of hippocampal neurons. The polyP-dependent neurotoxic nature of the TDP43A90V mutation was further confirmed as neutralization of polyP in ACM derived from mutant TDP43 astrocytes prevented MN death. Our results establish that human astrocytes carrying the TDP43A90V mutation exhibit a cell-autonomous pathological signature, hence providing an experimental model to decipher the molecular mechanisms underlying the generation of the neurotoxic phenotype.

2.
Hum Genet ; 142(8): 1263-1270, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37085629

RESUMO

Exocytosis is the process by which secretory vesicles fuse with the plasma membrane to deliver materials to the cell surface or to release cargoes to the extracellular space. The exocyst-an evolutionarily conserved octameric protein complex-mediates spatiotemporal control of SNARE complex assembly for vesicle fusion and tethering the secretory vesicles to the plasma membrane. The exocyst participates in diverse cellular functions, including protein trafficking to the plasma membrane, membrane extension, cell polarity, neurite outgrowth, ciliogenesis, cytokinesis, cell migration, autophagy, host defense, and tumorigenesis. Exocyst subunits are essential for cell viability; and mutations or variants in several exocyst subunits have been implicated in human diseases, mostly neurodevelopmental disorders and ciliopathies. These conditions often share common features such as developmental delay, intellectual disability, and brain abnormalities. In this review, we summarize the mutations and variants in exocyst subunits that have been linked to disease and discuss the implications of exocyst dysfunction in other disorders.


Assuntos
Doenças do Sistema Nervoso , Proteínas de Transporte Vesicular , Humanos , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Citoplasma/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Exocitose/genética , Doenças do Sistema Nervoso/genética
3.
Cell ; 186(4): 786-802.e28, 2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36754049

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that results from many diverse genetic causes. Although therapeutics specifically targeting known causal mutations may rescue individual types of ALS, these approaches cannot treat most cases since they have unknown genetic etiology. Thus, there is a pressing need for therapeutic strategies that rescue multiple forms of ALS. Here, we show that pharmacological inhibition of PIKFYVE kinase activates an unconventional protein clearance mechanism involving exocytosis of aggregation-prone proteins. Reducing PIKFYVE activity ameliorates ALS pathology and extends survival of animal models and patient-derived motor neurons representing diverse forms of ALS including C9ORF72, TARDBP, FUS, and sporadic. These findings highlight a potential approach for mitigating ALS pathogenesis that does not require stimulating macroautophagy or the ubiquitin-proteosome system.


Assuntos
Esclerose Lateral Amiotrófica , Fosfatidilinositol 3-Quinases , Animais , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Neurônios Motores , Mutação , Proteína FUS de Ligação a RNA/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Modelos Animais de Doenças
4.
Nat Commun ; 13(1): 6286, 2022 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-36271076

RESUMO

A GGGGCC24+ hexanucleotide repeat expansion (HRE) in the C9ORF72 gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), fatal neurodegenerative diseases with no cure or approved treatments that substantially slow disease progression or extend survival. Mechanistic underpinnings of neuronal death include C9ORF72 haploinsufficiency, sequestration of RNA-binding proteins in the nucleus, and production of dipeptide repeat proteins. Here, we used an adeno-associated viral vector system to deliver CRISPR/Cas9 gene-editing machineries to effectuate the removal of the HRE from the C9ORF72 genomic locus. We demonstrate successful excision of the HRE in primary cortical neurons and brains of three mouse models containing the expansion (500-600 repeats) as well as in patient-derived iPSC motor neurons and brain organoids (450 repeats). This resulted in a reduction of RNA foci, poly-dipeptides and haploinsufficiency, major hallmarks of C9-ALS/FTD, making this a promising therapeutic approach to these diseases.


Assuntos
Esclerose Lateral Amiotrófica , Demência Frontotemporal , Animais , Camundongos , Demência Frontotemporal/genética , Demência Frontotemporal/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Expansão das Repetições de DNA/genética , Sistemas CRISPR-Cas , Neurônios Motores/metabolismo , Dipeptídeos/metabolismo , RNA/metabolismo
5.
Nat Commun ; 13(1): 2776, 2022 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-35589706

RESUMO

Toxic dipeptide-repeat (DPR) proteins are produced from expanded G4C2 repeats in the C9ORF72 gene, the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Two DPR proteins, poly-PR and poly-GR, repress cellular translation but the molecular mechanism remains unknown. Here we show that poly-PR and poly-GR of ≥20 repeats inhibit the ribosome's peptidyl-transferase activity at nanomolar concentrations, comparable to specific translation inhibitors. High-resolution cryogenic electron microscopy (cryo-EM) reveals that poly-PR and poly-GR block the polypeptide tunnel of the ribosome, extending into the peptidyl-transferase center (PTC). Consistent with these findings, the macrolide erythromycin, which binds in the tunnel, competes with poly-PR and restores peptidyl-transferase activity. Our results demonstrate that strong and specific binding of poly-PR and poly-GR in the ribosomal tunnel blocks translation, revealing the structural basis of their toxicity in C9ORF72-ALS/FTD.


Assuntos
Esclerose Lateral Amiotrófica , Demência Frontotemporal , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Microscopia Crioeletrônica , Dipeptídeos/metabolismo , Demência Frontotemporal/genética , Demência Frontotemporal/metabolismo , Humanos , Proteínas/genética , Proteínas/metabolismo , Ribossomos/metabolismo , Transferases
6.
Neuron ; 110(10): 1656-1670.e12, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35276083

RESUMO

Non-cell-autonomous mechanisms contribute to neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), in which astrocytes release unidentified factors that are toxic to motoneurons (MNs). We report here that mouse and patient iPSC-derived astrocytes with diverse ALS/FTD-linked mutations (SOD1, TARDBP, and C9ORF72) display elevated levels of intracellular inorganic polyphosphate (polyP), a ubiquitous, negatively charged biopolymer. PolyP levels are also increased in astrocyte-conditioned media (ACM) from ALS/FTD astrocytes. ACM-mediated MN death is prevented by degrading or neutralizing polyP in ALS/FTD astrocytes or ACM. Studies further reveal that postmortem familial and sporadic ALS spinal cord sections display enriched polyP staining signals and that ALS cerebrospinal fluid (CSF) exhibits increased polyP concentrations. Our in vitro results establish excessive astrocyte-derived polyP as a critical factor in non-cell-autonomous MN degeneration and a potential therapeutic target for ALS/FTD. The CSF data indicate that polyP might serve as a new biomarker for ALS/FTD.


Assuntos
Esclerose Lateral Amiotrófica , Demência Frontotemporal , Esclerose Lateral Amiotrófica/genética , Animais , Astrócitos , Proteína C9orf72/genética , Meios de Cultivo Condicionados/farmacologia , Demência Frontotemporal/genética , Humanos , Camundongos , Neurônios Motores , Polifosfatos
8.
Cell ; 184(3): 689-708.e20, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33482083

RESUMO

The most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is a GGGGCC repeat expansion in the C9orf72 gene. We developed a platform to interrogate the chromatin accessibility landscape and transcriptional program within neurons during degeneration. We provide evidence that neurons expressing the dipeptide repeat protein poly(proline-arginine), translated from the C9orf72 repeat expansion, activate a highly specific transcriptional program, exemplified by a single transcription factor, p53. Ablating p53 in mice completely rescued neurons from degeneration and markedly increased survival in a C9orf72 mouse model. p53 reduction also rescued axonal degeneration caused by poly(glycine-arginine), increased survival of C9orf72 ALS/FTD-patient-induced pluripotent stem cell (iPSC)-derived motor neurons, and mitigated neurodegeneration in a C9orf72 fly model. We show that p53 activates a downstream transcriptional program, including Puma, which drives neurodegeneration. These data demonstrate a neurodegenerative mechanism dynamically regulated through transcription-factor-binding events and provide a framework to apply chromatin accessibility and transcription program profiles to neurodegeneration.


Assuntos
Proteína C9orf72/metabolismo , Expansão das Repetições de DNA/genética , Degeneração Neural/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Axônios/metabolismo , Proteína C9orf72/genética , Morte Celular , Células Cultivadas , Córtex Cerebral/patologia , Cromatina/metabolismo , Dano ao DNA , Modelos Animais de Doenças , Drosophila , Camundongos Endogâmicos C57BL , Degeneração Neural/patologia , Estabilidade Proteica , Transcrição Gênica , Proteínas Supressoras de Tumor/metabolismo
9.
Sci Rep ; 10(1): 14221, 2020 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-32848189

RESUMO

Mutations in CHMP2B, encoding a protein in the endosomal sorting complexes required for transport (ESCRT) machinery, causes frontotemporal dementia linked to chromosome 3 (FTD3). FTD, the second most common form of pre-senile dementia, can also be caused by genetic mutations in other genes, including TANK-binding kinase 1 (TBK1). How FTD-causing disease genes interact is largely unknown. We found that partial loss function of Ik2, the fly homologue of TBK1 also known as I-kappaB kinase ε (IKKε), enhanced the toxicity of mutant CHMP2B in the fly eye and that Ik2 overexpression suppressed the effect of mutant CHMP2B in neurons. Partial loss of function of Spn-F, a downstream phosphorylation target of Ik2, greatly enhanced the mutant CHMP2B phenotype. An interactome analysis to understand cellular processes regulated by Spn-F identified a network of interacting proteins including Spn-F, Ik2, dynein light chain, and Hook, an adaptor protein in early endosome transport. Partial loss of function of dynein light chain or Hook also enhanced mutant CHMP2B toxicity. These findings identify several evolutionarily conserved genes, including ik2/TBK1, cut up (encoding dynein light chain) and hook, as genetic modifiers of FTD3-associated mutant CHMP2B toxicity and implicate early endosome transport as a potential contributing pathway in FTD.


Assuntos
Proteínas de Drosophila/genética , Endossomos/fisiologia , Demência Frontotemporal/genética , Proteínas de Transporte Vesicular/genética , Animais , Modelos Animais de Doenças , Drosophila , Dineínas/genética , Quinase I-kappa B/genética , Proteínas Associadas aos Microtúbulos/genética
10.
Hum Mol Genet ; 27(8): 1382-1395, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29432529

RESUMO

Frontotemporal dementia (FTD) is one of the most prevalent forms of early-onset dementia. However, the pathological mechanisms driving neuronal atrophy in FTD remain poorly understood. Here we identify a conserved role for the novel pro-apoptotic protein plenty of SH3s (POSH)/SH3 domain containing ring finger 1 in mediating neuropathology in Drosophila and mammalian models of charged multivesicular body protein 2B (CHMP2BIntron5) associated FTD. Aberrant, AKT dependent, accumulation of POSH was observed throughout the nervous system of both Drosophila and mice expressing CHMP2BIntron5. Knockdown of POSH was shown to be neuroprotective and sufficient to alleviate aberrant neuronal morphology, behavioral deficits and premature-lethality in Drosophila models, as well as dendritic collapse and cell death in CHMP2BIntron5expressing rat primary neurons. POSH knockdown also ameliorated elevated markers of Jun N-terminal kinase and apoptotic cascades in both Drosophila and mammalian models. This study provides the first characterization of POSH as a potential component of an FTD neuropathology, identifying a novel apoptotic pathway with relevance to the FTD spectrum.


Assuntos
Proteínas de Transporte/genética , Proteínas do Citoesqueleto/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Demência Frontotemporal/genética , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Proteínas de Transporte Vesicular/genética , Animais , Animais Geneticamente Modificados , Apoptose/genética , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Proteínas do Citoesqueleto/antagonistas & inibidores , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Demência Frontotemporal/metabolismo , Demência Frontotemporal/patologia , Regulação da Expressão Gênica , Humanos , Íntrons , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Larva/genética , Larva/metabolismo , Longevidade/genética , Camundongos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Sistema Nervoso/metabolismo , Sistema Nervoso/patologia , Neurônios/patologia , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Transdução de Sinais , Proteínas de Transporte Vesicular/metabolismo
11.
Mol Neurodegener ; 12(1): 55, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28743268

RESUMO

Haploinsufficiency of GRN, the gene encoding progranulin (PGRN), causes frontotemporal lobar degeneration (FTLD), the second most common cause of early-onset dementia. Receptor-mediated lysosomal targeting has been shown to regulate brain PGRN levels, and complete deficiency of PGRN is a direct cause of neuronal ceroid lipofuscinosis (NCL), a lysosomal storage disease. Here we show that the lysosomal cysteine protease cathepsin L (Cat L) can mediate the proteolytic cleavage of intracellular PGRN into poly-granulin and granulin fragments. Further, PGRN and Cat L co-localize in lysosomes of HEK293 cells, iPSC-derived neurons and human cortical neurons from human postmortem tissue. These data identify Cat L as a key intracellular lysosomal PGRN protease, and provides an intriguing new link between lysosomal dysfunction and FTLD.


Assuntos
Catepsina L/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lisossomos/metabolismo , Proteínas/metabolismo , Células Cultivadas , Degeneração Lobar Frontotemporal/metabolismo , Humanos , Neurônios/metabolismo , Progranulinas
12.
Neuron ; 92(2): 383-391, 2016 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-27720481

RESUMO

GGGGCC repeat expansions in C9ORF72 are the most common genetic cause of both ALS and FTD. To uncover underlying pathogenic mechanisms, we found that DNA damage was greater, in an age-dependent manner, in motor neurons differentiated from iPSCs of multiple C9ORF72 patients than control neurons. Ectopic expression of the dipeptide repeat (DPR) protein (GR)80 in iPSC-derived control neurons increased DNA damage, suggesting poly(GR) contributes to DNA damage in aged C9ORF72 neurons. Oxidative stress was also increased in C9ORF72 neurons in an age-dependent manner. Pharmacological or genetic reduction of oxidative stress partially rescued DNA damage in C9ORF72 neurons and control neurons expressing (GR)80 or (GR)80-induced cellular toxicity in flies. Moreover, interactome analysis revealed that (GR)80 preferentially bound to mitochondrial ribosomal proteins and caused mitochondrial dysfunction. Thus, poly(GR) in C9ORF72 neurons compromises mitochondrial function and causes DNA damage in part by increasing oxidative stress, revealing another pathogenic mechanism in C9ORF72-related ALS and FTD.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Dano ao DNA , Demência Frontotemporal/metabolismo , Mitocôndrias/metabolismo , Neurônios Motores/metabolismo , Estresse Oxidativo/genética , Esclerose Lateral Amiotrófica/genética , Arginina/metabolismo , Western Blotting , Proteína C9orf72 , Linhagem Celular , Expansão das Repetições de DNA , Dipeptídeos/metabolismo , Demência Frontotemporal/genética , Glicina/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas , Proteínas/genética , Proteína Supressora de Tumor p53/metabolismo
13.
Hum Mol Genet ; 23(6): 1467-78, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24163244

RESUMO

Progranulin (GRN) mutations causing haploinsufficiency are a major cause of frontotemporal lobar degeneration (FTLD-TDP). Recent discoveries demonstrating sortilin (SORT1) is a neuronal receptor for PGRN endocytosis and a determinant of plasma PGRN levels portend the development of enhancers targeting the SORT1-PGRN axis. We demonstrate the preclinical efficacy of several approaches through which impairing PGRN's interaction with SORT1 restores extracellular PGRN levels. Our report is the first to demonstrate the efficacy of enhancing PGRN levels in iPSC neurons derived from frontotemporal dementia (FTD) patients with PGRN deficiency. We validate a small molecule preferentially increases extracellular PGRN by reducing SORT1 levels in various mammalian cell lines and patient-derived iPSC neurons and lymphocytes. We further demonstrate that SORT1 antagonists and a small-molecule binder of PGRN588₋593, residues critical for PGRN-SORT1 binding, inhibit SORT1-mediated PGRN endocytosis. Collectively, our data demonstrate that the SORT1-PGRN axis is a viable target for PGRN-based therapy, particularly in FTD-GRN patients.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Endocitose/efeitos dos fármacos , Demência Frontotemporal/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Piridinas/farmacologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Linhagem Celular Tumoral , Demência Frontotemporal/patologia , Variação Genética , Células HEK293 , Haploinsuficiência , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Linfócitos/metabolismo , Progranulinas , Reprodutibilidade dos Testes
14.
FASEB J ; 28(2): 667-75, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24158394

RESUMO

Endosomal sorting complexes required for transport (ESCRTs) mediate sorting of ubiquitinated membrane proteins into multivesicular bodies en route to lysosomes for degradation. A mutation in CHMP2B (CHMP2B(Intron5), an ESCRT-III component) that is associated with a hereditary form of frontotemporal dementia (FTD3) disrupts the endosomal-lysosomal pathway and causes accumulation of autophagosomes and multilamellar structures. We previously demonstrated that expression of CHMP2B(Intron5) in the Drosophila eye using GMR-Gal4 causes misregulation of the Toll receptor pathway. Here, we show that ectopic expression of CHMP2B(Intron5) using eyeless-Gal4 (ey>CHMP2B(Intron5)), a driver with different spatiotemporal expression attributes than GMR-Gal4 in the Drosophila eye, causes eye deformities when compared to expression of wild-type CHMP2B (CHMP2B(WT)) and the Drosophila homologue of CHMP2B (CG4618). In addition, ey>CHMP2B(Intron5) flies showed defects in photoreceptor cell patterning and phototactic behavior. Furthermore, ey>CHMP2B(Intron5) flies showed accumulation of Notch in enlarged endosomes and up-regulation of Notch activity. Partial loss of Notch activity in ey>CHMP2B(Intron5) flies significantly rescued eye deformities, photoreceptor patterning defect, and phototactic behavior defect, indicating that these defects are primarily due to Notch misregulation. These results demonstrate that CHMP2B(Intron5) preferentially affects different receptor signaling pathways in a cellular and developmental context-dependent manner.


Assuntos
Proteínas de Drosophila/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Olho/metabolismo , Olho/patologia , Demência Frontotemporal/metabolismo , Receptores Notch/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animais , Western Blotting , Drosophila , Proteínas de Drosophila/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Demência Frontotemporal/genética , Reação em Cadeia da Polimerase , Receptores Notch/genética , Proteínas de Transporte Vesicular/genética
15.
J Neurosci ; 33(12): 5352-61, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-23516300

RESUMO

Frontotemporal dementia (FTD) is a neurodegenerative disease with hallmark deficits in social and emotional function. Heterozygous loss-of-function mutations in GRN, the progranulin gene, are a common genetic cause of the disorder, but the mechanisms by which progranulin haploinsufficiency causes neuronal dysfunction in FTD are unclear. Homozygous progranulin knock-out (Grn(-/-)) mice have been studied as a model of this disorder and show behavioral deficits and a neuroinflammatory phenotype with robust microglial activation. However, homozygous GRN mutations causing complete progranulin deficiency were recently shown to cause a different neurological disorder, neuronal ceroid lipofuscinosis, suggesting that the total absence of progranulin may have effects distinct from those of haploinsufficiency. Here, we studied progranulin heterozygous (Grn(+/-)) mice, which model progranulin haploinsufficiency. We found that Grn(+/-) mice developed age-dependent social and emotional deficits potentially relevant to FTD. However, unlike Grn(-/-) mice, behavioral deficits in Grn(+/-) mice occurred in the absence of gliosis or increased expression of tumor necrosis factor-α. Instead, we found neuronal abnormalities in the amygdala, an area of selective vulnerability in FTD, in Grn(+/-) mice. Our findings indicate that FTD-related deficits resulting from progranulin haploinsufficiency can develop in the absence of detectable gliosis and neuroinflammation, thereby dissociating microglial activation from functional deficits and suggesting an important effect of progranulin deficiency on neurons.


Assuntos
Encefalite , Demência Frontotemporal/genética , Demência Frontotemporal/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Tonsila do Cerebelo/patologia , Tonsila do Cerebelo/fisiopatologia , Animais , Comportamento Animal/fisiologia , Condicionamento Psicológico/fisiologia , Modelos Animais de Doenças , Emoções/fisiologia , Feminino , Demência Frontotemporal/patologia , Gliose , Granulinas , Haploinsuficiência/fisiologia , Homozigoto , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/patologia , Microglia/fisiologia , Fenótipo , Progranulinas , Comportamento Social , Comportamento Espacial/fisiologia
16.
Methods Mol Biol ; 916: 387-402, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22914955

RESUMO

Human embryonic stem cells and induced pluripotent stem cells offer great hope for studies of pathogenic mechanisms of disease and cell-based therapies. One powerful approach to manipulate the behaviors of human stem cells and their progenies is through microRNAs (miRNAs), a class of small noncoding RNAs that regulate gene expression at the posttranscriptional level. Each miRNA may target up to hundreds of mRNAs; some are specifically expressed in progenitor cells and affect multiple cellular processes. Here we present experimental protocols for investigating the endogenous functions of specific miRNAs in the proliferation, survival, and migration of human neural progenitor cells derived from embryonic stem cells. These methods may be applicable to protein factors and neural progenitor cells derived from patient-specific induced pluripotent stem cells.


Assuntos
MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Animais , Encéfalo/citologia , Encéfalo/embriologia , Bromodesoxiuridina/metabolismo , Diferenciação Celular , Linhagem Celular , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Feminino , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Gravidez , Coloração e Rotulagem , Transplante de Células-Tronco , Transfecção
18.
ChemMedChem ; 7(4): 571-7, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22354575

RESUMO

SecA is a central component of the general secretion system that is essential for bacterial growth and thus an ideal target for antimicrobial agents. A series of fluorescein analogues were first screened against the ATPase activity using the truncated unregulated SecA catalytic domain. Rose bengal (RB) and erythrosin B (EB) were found to be potent inhibitors SecA with IC(50) values of 0.5 µM and 2 µM, respectively. RB and EB inhibit the catalytic SecA ATPase more effectively than the F(1) F(0) -proton ATPase. We used three assays to test the effect of these compounds on full-length SecA ATPase: in solution (intrinsic ATPase), in membrane preparation, and translocation ATPase. RB and EB show the following trend in terms of IC(50) values: translocation ATPase

Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Proteínas de Bactérias/antagonistas & inibidores , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Eritrosina/farmacologia , Fluoresceína/química , Rosa Bengala/farmacologia , Antibacterianos/química , Antibacterianos/farmacologia , Bacillus subtilis/efeitos dos fármacos , Bacillus subtilis/enzimologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Eritrosina/química , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Concentração Inibidora 50 , Proteínas de Membrana Transportadoras , Modelos Moleculares , Transporte Proteico/efeitos dos fármacos , Rosa Bengala/química , Canais de Translocação SEC , Proteínas SecA
19.
PLoS One ; 6(10): e26454, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22028881

RESUMO

The reduced production or activity of the cysteine-rich glycoprotein progranulin is responsible for about 20% of cases of familial frontotemporal dementia. However, little is known about the molecular mechanisms that govern the level and secretion of progranulin. Here we show that progranulin is expressed in mouse cortical neurons and more prominently in mouse microglia in culture and is abundant in the endoplasmic reticulum (ER) and Golgi. Using chemical crosslinking, immunoprecipitation, and mass spectrometry, we found that progranulin is bound to a network of ER Ca(2+)-binding chaperones including BiP, calreticulin, GRP94, and four members of the protein disulfide isomerase (PDI) family. Loss of ERp57 inhibits progranulin secretion. Thus, progranulin is a novel substrate of several PDI family proteins and modulation of the ER chaperone network may be a therapeutic target for controlling progranulin secretion.


Assuntos
Demência Frontotemporal/metabolismo , Glicoproteínas/deficiência , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Animais , Biomarcadores/metabolismo , Córtex Cerebral/citologia , Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica , Granulinas , Células HEK293 , Humanos , Camundongos , Microglia/metabolismo , Chaperonas Moleculares/metabolismo , Neurônios/metabolismo , Progranulinas , Ligação Proteica , Transporte Proteico
20.
Nat Cell Biol ; 13(10): 1214-23, 2011 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-21909099

RESUMO

The midbody is a singular organelle formed between daughter cells during cytokinesis and required for their final separation. Midbodies persist in cells long after division as midbody derivatives (MB(d)s), but their fate is unclear. Here we show that MB(d)s are inherited asymmetrically by the daughter cell with the older centrosome. They selectively accumulate in stem cells, induced pluripotent stem cells and potential cancer 'stem cells' in vivo and in vitro. MB(d) loss accompanies stem-cell differentiation, and involves autophagic degradation mediated by binding of the autophagic receptor NBR1 to the midbody protein CEP55. Differentiating cells and normal dividing cells do not accumulate MB(d)s and possess high autophagic activity. Stem cells and cancer cells accumulate MB(d)s by evading autophagosome encapsulation and exhibit low autophagic activity. MB(d) enrichment enhances reprogramming to induced pluripotent stem cells and increases the in vitro tumorigenicity of cancer cells. These results indicate unexpected roles for MB(d)s in stem cells and cancer 'stem cells'.


Assuntos
Autofagia , Transformação Celular Neoplásica/patologia , Reprogramação Celular , Células-Tronco Embrionárias/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Neoplásicas/patologia , Organelas/patologia , Animais , Autofagia/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Divisão Celular , Linhagem Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Centrossomo/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Técnicas de Cocultura , Células-Tronco Embrionárias/metabolismo , Células HeLa , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Lisossomos/metabolismo , Camundongos , Células-Tronco Neoplásicas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Organelas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA