Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros













Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(10): e2313540121, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38416681

RESUMO

Mutations in PTEN-induced putative kinase 1 (PINK1) cause autosomal recessive early-onset Parkinson's disease (PD). PINK1 is a Ser/Thr kinase that regulates mitochondrial quality control by triggering mitophagy mediated by the ubiquitin (Ub) ligase Parkin. Upon mitochondrial damage, PINK1 accumulates on the outer mitochondrial membrane forming a high-molecular-weight complex with the translocase of the outer membrane (TOM). PINK1 then phosphorylates Ub, which enables recruitment and activation of Parkin followed by autophagic clearance of the damaged mitochondrion. Thus, Parkin-dependent mitophagy hinges on the stable accumulation of PINK1 on the TOM complex. Yet, the mechanism linking mitochondrial stressors to PINK1 accumulation and whether the translocases of the inner membrane (TIMs) are also involved remain unclear. Herein, we demonstrate that mitochondrial stress induces the formation of a PINK1-TOM-TIM23 supercomplex in human cultured cell lines, dopamine neurons, and midbrain organoids. Moreover, we show that PINK1 is required to stably tether the TOM to TIM23 complexes in response to stress such that the supercomplex fails to accumulate in cells lacking PINK1. This tethering is dependent on an interaction between the PINK1 N-terminal-C-terminal extension module and the cytosolic domain of the Tom20 subunit of the TOM complex, the disruption of which, by either designer or PD-associated PINK1 mutations, inhibits downstream mitophagy. Together, the findings provide key insight into how PINK1 interfaces with the mitochondrial import machinery, with important implications for the mechanisms of mitochondrial quality control and PD pathogenesis.


Assuntos
Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Proteínas Quinases , Humanos , Proteínas de Transporte/metabolismo , Mitocôndrias/metabolismo , Fosforilação , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
2.
Brain ; 146(5): 1859-1872, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-36370000

RESUMO

The association between glucocerebrosidase, encoded by GBA, and Parkinson's disease (PD) highlights the role of the lysosome in PD pathogenesis. Genome-wide association studies in PD have revealed multiple associated loci, including the GALC locus on chromosome 14. GALC encodes the lysosomal enzyme galactosylceramidase, which plays a pivotal role in the glycosphingolipid metabolism pathway. It is still unclear whether GALC is the gene driving the association in the chromosome 14 locus and, if so, by which mechanism. We first aimed to examine whether variants in the GALC locus and across the genome are associated with galactosylceramidase activity. We performed a genome-wide association study in two independent cohorts from (i) Columbia University; and (ii) the Parkinson's Progression Markers Initiative study, followed by a meta-analysis with a total of 976 PD patients and 478 controls with available data on galactosylceramidase activity. We further analysed the effects of common GALC variants on expression and galactosylceramidase activity using genomic colocalization methods. Mendelian randomization was used to study whether galactosylceramidase activity may be causal in PD. To study the role of rare GALC variants, we analysed sequencing data from 5028 PD patients and 5422 controls. Additionally, we studied the functional impact of GALC knockout on alpha-synuclein accumulation and on glucocerebrosidase activity in neuronal cell models and performed in silico structural analysis of common GALC variants associated with altered galactosylceramidase activity. The top hit in PD genome-wide association study in the GALC locus, rs979812, is associated with increased galactosylceramidase activity (b = 1.2; SE = 0.06; P = 5.10 × 10-95). No other variants outside the GALC locus were associated with galactosylceramidase activity. Colocalization analysis demonstrated that rs979812 was also associated with increased galactosylceramidase expression. Mendelian randomization suggested that increased galactosylceramidase activity may be causally associated with PD (b = 0.025, SE = 0.007, P = 0.0008). We did not find an association between rare GALC variants and PD. GALC knockout using CRISPR-Cas9 did not lead to alpha-synuclein accumulation, further supporting that increased rather than reduced galactosylceramidase levels may be associated with PD. The structural analysis demonstrated that the common variant p.I562T may lead to improper maturation of galactosylceramidase affecting its activity. Our results nominate GALC as the gene associated with PD in this locus and suggest that the association of variants in the GALC locus may be driven by their effect of increasing galactosylceramidase expression and activity. Whether altering galactosylceramidase activity could be considered as a therapeutic target should be further studied.


Assuntos
Doença de Parkinson , Humanos , Doença de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Galactosilceramidase/genética , Galactosilceramidase/metabolismo , Glucosilceramidase/genética , Estudo de Associação Genômica Ampla , Mutação , Hidrolases/genética
3.
Sci Rep ; 12(1): 17176, 2022 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-36229560

RESUMO

The use of human derived induced pluripotent stem cells (hiPSCs) differentiated to dopaminergic (DA) neurons offers a valuable experimental model to decorticate the cellular and molecular mechanisms of Parkinson's disease (PD) pathogenesis. However, the existing approaches present with several limitations, notably the lengthy time course of the protocols and the high variability in the yield of DA neurons. Here we report on the development of an improved approach that combines neurogenin-2 programming with the use of commercially available midbrain differentiation kits for a rapid, efficient, and reproducible directed differentiation of hiPSCs to mature and functional induced DA (iDA) neurons, with minimum contamination by other brain cell types. Gene expression analysis, associated with functional characterization examining neurotransmitter release and electrical recordings, support the functional identity of the iDA neurons to A9 midbrain neurons. iDA neurons showed selective vulnerability when exposed to 6-hydroxydopamine, thus providing a viable in vitro approach for modeling PD and for the screening of small molecules with neuroprotective proprieties.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença de Parkinson , Diferenciação Celular/genética , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Humanos , Mesencéfalo/metabolismo , Neurotransmissores/metabolismo , Oxidopamina/metabolismo , Oxidopamina/farmacologia , Doença de Parkinson/metabolismo
4.
Cells ; 11(13)2022 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-35805181

RESUMO

The best-known hallmarks of Parkinson's disease (PD) are the motor deficits that result from the degeneration of dopaminergic neurons in the substantia nigra. Dopaminergic neurons are thought to be particularly susceptible to mitochondrial dysfunction. As such, for their survival, they rely on the elaborate quality control mechanisms that have evolved in mammalian cells to monitor mitochondrial function and eliminate dysfunctional mitochondria. Mitophagy is a specialized type of autophagy that mediates the selective removal of damaged mitochondria from cells, with the net effect of dampening the toxicity arising from these dysfunctional organelles. Despite an increasing understanding of the molecular mechanisms that regulate the removal of damaged mitochondria, the detailed molecular link to PD pathophysiology is still not entirely clear. Herein, we review the fundamental molecular pathways involved in PINK1/Parkin-mediated and receptor-mediated mitophagy, the evidence for the dysfunction of these pathways in PD, and recently-developed state-of-the art assays for measuring mitophagy in vitro and in vivo.


Assuntos
Mitofagia , Doença de Parkinson , Animais , Autofagia/fisiologia , Mamíferos/metabolismo , Mitocôndrias/metabolismo , Mitofagia/fisiologia , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Proteínas Quinases/metabolismo
5.
Cell Rep ; 40(3): 111102, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35858558

RESUMO

The nervous system spread of alpha-synuclein fibrils is thought to cause Parkinson's disease (PD) and other synucleinopathies; however, the mechanisms underlying internalization and cellular spread are enigmatic. Here, we use confocal and superresolution microscopy, subcellular fractionation, and electron microscopy (EM) of immunogold-labeled α-synuclein preformed fibrils (PFFs) to demonstrate that this form of the protein undergoes rapid internalization and is targeted directly to lysosomes in as little as 2 min. Uptake of PFFs is disrupted by macropinocytic inhibitors and circumvents classical endosomal pathways. Immunogold-labeled PFFs are seen at the highly curved inward edge of membrane ruffles, in newly formed macropinosomes, in multivesicular bodies and in lysosomes. While most fibrils remain in lysosomes, a portion is transferred to neighboring naive cells along with markers of exosomes. These data indicate that PFFs use a unique internalization mechanism as a component of cell-to-cell propagation.


Assuntos
Doença de Parkinson , Sinucleinopatias , Endossomos/metabolismo , Humanos , Lisossomos/metabolismo , Doença de Parkinson/metabolismo , alfa-Sinucleína/metabolismo
6.
Curr Protein Pept Sci ; 23(3): 129-132, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35598241

RESUMO

One salient hallmark of neurodegeneration is the accumulation of toxic protein aggregates in neuronal cells. This proteotoxicity culminates in the deterioration of neuronal function. In AD and related tauopathies, the microtubule-associated protein tau becomes hyperphosphorylated. Hyperphosphorylated tau forms neurofibrillary tangles (NFTs) within neurons, which constitute a unique feature of tauopathies, including AD. A recent study has exploited a novel molecular strategy to counteract hyperphosphorylated tau and enhance its degradation. Analogous to the PROTAC methodology, a novel dephosphorylation targeting chimera (DEPTAC) was designed to promote the molecular interaction between tau and phosphatase, which, in turn, augments its degradation. Herein, we briefly discuss this novel finding and its potential therapeutic implications.


Assuntos
Doença de Alzheimer , Tauopatias , Doença de Alzheimer/metabolismo , Humanos , Emaranhados Neurofibrilares/metabolismo , Neurônios/metabolismo , Fosforilação , Tauopatias/genética , Tauopatias/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
7.
Bioessays ; 44(6): e2200008, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35417040

RESUMO

Selective protein degradation maintains cellular homeostasis, but this process is disrupted in many diseases. Targeted protein degradation (TPD) approaches, built upon existing cellular mechanisms, are promising methods for therapeutically regulating protein levels. Here, we review the diverse palette of tools that are now available for doing so throughout the gene expression pathway and in specific cellular compartments. These include methods for directly removing targeted proteins via the ubiquitin proteasome system with proteolysis targeting chimeras (PROTACs) or dephosphorylation targeting chimeras (DEPTACs). Similar effects can also be achieved through the lysosomal system with autophagy-targeting chimeras (AUTACs), autophagosome tethering compounds (ATTECs), and lysosome targeting chimeras (LYTACs). Other methods act upstream to degrade RNAs (ribonuclease targeting chimeras; RIBOTACs) or transcription factors (transcription factor targeting chimeras; TRAFTACs), offering control throughout the gene expression process. We highlight the evolution and function of these methods and discuss their clinical implications in diverse disease contexts.


Assuntos
Lisossomos , Complexo de Endopeptidases do Proteassoma , Autofagia , Lisossomos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Fatores de Transcrição/metabolismo
8.
Bioessays ; 43(2): e2000212, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33210303

RESUMO

Autophagy functions in both selective and non-selective ways to maintain cellular homeostasis. Endoplasmic reticulum autophagy (ER-phagy) is a subclass of autophagy responsible for the degradation of the endoplasmic reticulum through selective encapsulation into autophagosomes. ER-phagy occurs both under physiological conditions and in response to stress cues, and plays a crucial role in maintaining the homeostatic control of the organelle. Although specific receptors that target parts of the ER membrane, as well as, internal proteins for lysosomal degradation have been identified, the molecular regulation of ER-phagy has been elusive. Recent work has uncovered novel regulators of ER-phagy that involve post-translational modifications of ER-resident proteins and functional cross-talk with other cellular processes. Herein, we discuss how morphology affects the function of the peripheral ER, and how ER-phagy modulates the turnover of this organelle. We also address how ER-phagy is regulated at the molecular level, considering implications relevant to human diseases.


Assuntos
Estresse do Retículo Endoplasmático , Proteínas de Membrana , Autofagia , Retículo Endoplasmático/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Processamento de Proteína Pós-Traducional
9.
Front Cell Neurosci ; 14: 594304, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33281561

RESUMO

Traumatic brain injury (TBI) is the leading cause of disability and mortality in children and young adults and has a profound impact on the socio-economic wellbeing of patients and their families. Initially, brain damage is caused by mechanical stress-induced axonal injury and vascular dysfunction, which can include hemorrhage, blood-brain barrier disruption, and ischemia. Subsequent neuronal degeneration, chronic inflammation, demyelination, oxidative stress, and the spread of excitotoxicity can further aggravate disease pathology. Thus, TBI treatment requires prompt intervention to protect against neuronal and vascular degeneration. Rapid advances in the field of stem cells (SCs) have revolutionized the prospect of repairing brain function following TBI. However, more than that, SCs can contribute substantially to our knowledge of this multifaced pathology. Research, based on human induced pluripotent SCs (hiPSCs) can help decode the molecular pathways of degeneration and recovery of neuronal and glial function, which makes these cells valuable tools for drug screening. Additionally, experimental approaches that include hiPSC-derived engineered tissues (brain organoids and bio-printed constructs) and biomaterials represent a step forward for the field of regenerative medicine since they provide a more suitable microenvironment that enhances cell survival and grafting success. In this review, we highlight the important role of hiPSCs in better understanding the molecular pathways of TBI-related pathology and in developing novel therapeutic approaches, building on where we are at present. We summarize some of the most relevant findings for regenerative therapies using biomaterials and outline key challenges for TBI treatments that remain to be addressed.

10.
Endocrinology ; 161(12)2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33159513

RESUMO

In mammals, protein degradation is mediated selectively by the ubiquitin proteasome system (UPS) and the autophagic-lysosomal system. Over the past decades, N-degron pathways have been shown to be responsible for the selective degradation of proteins that harbor destabilizing N-terminal motifs. Recent studies have employed these pathways in the development of proteolysis targeting chimeras (PROTACs) composed of a degradation module linked to a substrate recognition domain to target proteins encoded by cancer-related genes for proteasomal destruction. Herein we provide an overview of PROTACs in the context of the N-degron concept and address the application of this technique to curb the migration and invasion of cancer cells, with a focus on the far-reaching potential of exploiting N-degron pathways for therapeutic purposes.


Assuntos
Autofagia/fisiologia , Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas/metabolismo , Proteólise , Humanos
11.
Commun Chem ; 3(1): 149, 2020 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-36703340
12.
Nat Cell Biol ; 20(7): 789-799, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29941930

RESUMO

Cellular senescence is a tumour suppressor programme characterized by a stable cell cycle arrest. Here we report that cellular senescence triggered by a variety of stimuli leads to diminished ribosome biogenesis and the accumulation of both rRNA precursors and ribosomal proteins. These defects were associated with reduced expression of several ribosome biogenesis factors, the knockdown of which was also sufficient to induce senescence. Genetic analysis revealed that Rb but not p53 was required for the senescence response to altered ribosome biogenesis. Mechanistically, the ribosomal protein S14 (RPS14 or uS11) accumulates in the soluble non-ribosomal fraction of senescent cells, where it binds and inhibits CDK4 (cyclin-dependent kinase 4). Overexpression of RPS14 is sufficient to inhibit Rb phosphorylation, inducing cell cycle arrest and senescence. Here we describe a mechanism for maintaining the senescent cell cycle arrest that may be relevant for cancer therapy, as well as biomarkers to identify senescent cells.


Assuntos
Pontos de Checagem do Ciclo Celular , Senescência Celular , Neoplasias/metabolismo , Proteína do Retinoblastoma/metabolismo , Ribossomos/metabolismo , Fatores de Coagulação Sanguínea/genética , Fatores de Coagulação Sanguínea/metabolismo , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Células HEK293 , Humanos , Neoplasias/genética , Neoplasias/patologia , Células PC-3 , Fosforilação , Ligação Proteica , Precursores de RNA/biossíntese , Precursores de RNA/genética , RNA Ribossômico/biossíntese , RNA Ribossômico/genética , Proteínas de Ligação a RNA , Proteína do Retinoblastoma/genética , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Ribossomos/genética , Transdução de Sinais , Fatores de Tempo
13.
Nucleic Acids Res ; 45(6): 3017-3030, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-27932455

RESUMO

Enhancers are intergenic DNA elements that regulate the transcription of target genes in response to signaling pathways by interacting with promoters over large genomic distances. Recent studies have revealed that enhancers are bi-directionally transcribed into enhancer RNAs (eRNAs). Using single-molecule fluorescence in situ hybridization (smFISH), we investigated the eRNA-mediated regulation of transcription during estrogen induction in MCF-7 cells. We demonstrate that eRNAs are localized exclusively in the nucleus and are induced with similar kinetics as target mRNAs. However, eRNAs are mostly nascent at enhancers and their steady-state levels remain lower than those of their cognate mRNAs. Surprisingly, at the single-allele level, eRNAs are rarely co-expressed with their target loci, demonstrating that active gene transcription does not require the continuous transcription of eRNAs or their accumulation at enhancers. When co-expressed, sub-diffraction distance measurements between nascent mRNA and eRNA signals reveal that co-transcription of eRNAs and mRNAs rarely occurs within closed enhancer-promoter loops. Lastly, basal eRNA transcription at enhancers, but not E2-induced transcription, is maintained upon depletion of MLL1 and ERα, suggesting some degree of chromatin accessibility prior to signal-dependent activation of transcription. Together, our findings suggest that eRNA accumulation at enhancer-promoter loops is not required to sustain target gene transcription.


Assuntos
Elementos Facilitadores Genéticos , Regiões Promotoras Genéticas , RNA não Traduzido/biossíntese , Transcrição Gênica , Estradiol/farmacologia , Receptor alfa de Estrogênio/fisiologia , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Histona-Lisina N-Metiltransferase/fisiologia , Humanos , Células MCF-7 , Modelos Moleculares , Proteína de Leucina Linfoide-Mieloide/fisiologia , RNA Mensageiro/biossíntese , RNA não Traduzido/fisiologia , Receptores Purinérgicos P2Y2/biossíntese , Receptores Purinérgicos P2Y2/genética , Análise de Célula Única
14.
Proc Natl Acad Sci U S A ; 112(13): E1587-93, 2015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25770220

RESUMO

Naive CD4 T cells differentiate into several effector lineages, which generate a stronger and more rapid response to previously encountered immunological challenges. Although effector function is a key feature of adaptive immunity, the molecular basis of this process is poorly understood. Here, we investigated the spatiotemporal regulation of cytokine gene expression in resting and restimulated effector T helper 1 (Th1) cells. We found that the Lymphotoxin (LT)/TNF alleles, which encode TNF-α, were closely juxtaposed shortly after T-cell receptor (TCR) engagement, when transcription factors are limiting. Allelic pairing required a nuclear myosin, myosin VI, which is rapidly recruited to the LT/TNF locus upon restimulation. Furthermore, transcription was paused at the TNF locus and other related genes in resting Th1 cells and released in a myosin VI-dependent manner following activation. We propose that homologous pairing and myosin VI-mediated transcriptional pause release account for the rapid and efficient expression of genes induced by an external stimulus.


Assuntos
Cadeias Pesadas de Miosina/fisiologia , Células Th1/metabolismo , Transcrição Gênica , Alelos , Animais , Núcleo Celular/metabolismo , Citocinas/metabolismo , Hibridização in Situ Fluorescente , Linfotoxina-alfa/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cadeias Pesadas de Miosina/genética , RNA Polimerase II/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/genética
15.
Mol Cell ; 54(1): 56-66, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24613343

RESUMO

Interchromosomal associations can regulate gene expression, but little is known about the molecular basis of such associations. In response to antigen stimulation, naive T cells can differentiate into Th1, Th2, and Th17 cells expressing IFN-γ, IL-4, and IL-17, respectively. We previously reported that in naive T cells, the IFN-γ locus is associated with the Th2 cytokine locus. Here we show that the Th2 locus additionally associates with the IL-17 locus. This association requires a DNase I hypersensitive region (RHS6) at the Th2 locus. RHS6 and the IL-17 promoter both bear Oct-1 binding sites. Deletion of either of these sites or Oct-1 gene impairs the association. Oct-1 and CTCF bind their cognate sites cooperatively, and CTCF deficiency similarly impairs the association. Finally, defects in the association lead to enhanced IL-17 induction. Collectively, our data indicate Th17 lineage differentiation is restrained by the Th2 locus via interchromosomal associations organized by Oct-1 and CTCF.


Assuntos
Cromossomos de Mamíferos , Interleucina-17/metabolismo , Fator 1 de Transcrição de Octâmero/metabolismo , Proteínas Repressoras/metabolismo , Células Th17/metabolismo , Células Th2/metabolismo , Animais , Sítios de Ligação , Fator de Ligação a CCCTC , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Desoxirribonuclease I/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Loci Gênicos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interleucina-17/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator 1 de Transcrição de Octâmero/deficiência , Fator 1 de Transcrição de Octâmero/genética , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Deleção de Sequência , Células Th17/imunologia , Células Th2/imunologia , Fatores de Tempo
16.
Yale J Biol Med ; 84(2): 113-6, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21698042

RESUMO

George Emil Palade's scientific contributions significantly advanced the field of modern cell biology. He pioneered a multidisciplinary approach, combining cell fractionation, biochemistry, and electron microscopy, which led to the identification of the ribosome as the site of protein synthesis and elucidated the eukaryotic secretory pathway. For these accomplishments, Palade, along with Albert Claude and Christian de Duve, won the 1974 Nobel Prize in Physiology or Medicine. This article provides an overview of Palade's seminal research in the context of the early developments in the field.


Assuntos
Biologia Celular/história , Bioquímica/história , Fracionamento Celular , História do Século XX , História do Século XXI , Microscopia Eletrônica , Prêmio Nobel , Biossíntese de Proteínas , Ribossomos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA