Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Hum Mol Genet ; 32(10): 1673-1682, 2023 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-36611007

RESUMO

The abnormal expansion of GGGGCC hexanucleotide repeats within the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The accumulation of GGGGCC repeat-containing RNAs as RNA foci, and the deposition of dipeptide repeat proteins (DPR) produced from these repeat RNAs by unconventional translation are major pathological hallmarks of C9orf72-linked ALS/FTD (C9-ALS/FTD), and are both thought to play a crucial role in the pathogenesis of these diseases. Because GGGGCC repeat RNA is likely to be the most upstream therapeutic target in the pathogenic cascade of C9-ALS/FTD, lowering the cellular level of GGGGCC repeat RNA is expected to mitigate repeat RNA toxicity, and will therefore be a disease-modifying therapeutic strategy for the treatment of C9-ALS/FTD. In this study, we demonstrated using a Drosophila model of C9-ALS/FTD that elevated expression of a subset of human RNA-binding proteins that bind to GGGGCC repeat RNA, including hnRNPA3, IGF2BP1, hnRNPA2B1, hnRNPR and SF3B3, reduces the level of GGGGCC repeat RNA, resulting in the suppression of neurodegeneration. We further showed that hnRNPA3-mediated reduction of GGGGCC repeat RNA suppresses disease pathology, such as RNA foci and DPR accumulation. These results demonstrate that hnRNPA3 and other RNA-binding proteins negatively regulate the level of GGGGCC repeat RNA, and mitigate repeat RNA toxicity in vivo, indicating the therapeutic potential of the repeat RNA-lowering approach mediated by endogenous RNA-binding proteins for the treatment of C9-ALS/FTD.


Assuntos
Esclerose Lateral Amiotrófica , Demência Frontotemporal , Doença de Pick , Animais , Humanos , Demência Frontotemporal/patologia , Esclerose Lateral Amiotrófica/metabolismo , RNA/genética , RNA/metabolismo , Proteína C9orf72/genética , Drosophila/genética , Drosophila/metabolismo , Doença de Pick/genética , Proteínas/genética , Dipeptídeos/genética , Expansão das Repetições de DNA/genética
2.
Acta Neuropathol ; 145(5): 573-595, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36939875

RESUMO

Lipid interaction with α-synuclein (αSyn) has been long implicated in the pathogenesis of Parkinson's disease (PD). However, it has not been fully determined which lipids are involved in the initiation of αSyn aggregation in PD. Here exploiting genetic understanding associating the loss-of-function mutation in Synaptojanin 1 (SYNJ1), a phosphoinositide phosphatase, with familial PD and analysis of postmortem PD brains, we identified a novel lipid molecule involved in the toxic conversion of αSyn and its relation to PD. We first established a SYNJ1 knockout cell model and found SYNJ1 depletion increases the accumulation of pathological αSyn. Lipidomic analysis revealed SYNJ1 depletion elevates the level of its substrate phosphatidylinositol-3,4,5-trisphosphate (PIP3). We then employed Caenorhabditis elegans model to examine the effect of SYNJ1 defect on the neurotoxicity of αSyn. Mutations in SYNJ1 accelerated the accumulation of αSyn aggregation and induced locomotory defects in the nematodes. These results indicate that functional loss of SYNJ1 promotes the pathological aggregation of αSyn via the dysregulation of its substrate PIP3, leading to the aggravation of αSyn-mediated neurodegeneration. Treatment of cultured cell line and primary neurons with PIP3 itself or with PIP3 phosphatase inhibitor resulted in intracellular formation of αSyn inclusions. Indeed, in vitro protein-lipid overlay assay validated that phosphoinositides, especially PIP3, strongly interact with αSyn. Furthermore, the aggregation assay revealed that PIP3 not only accelerates the fibrillation of αSyn, but also induces the formation of fibrils sharing conformational and biochemical characteristics similar to the fibrils amplified from the brains of PD patients. Notably, the immunohistochemical and lipidomic analyses on postmortem brain of patients with sporadic PD showed increased PIP3 level and its colocalization with αSyn. Taken together, PIP3 dysregulation promotes the pathological aggregation of αSyn and increases the risk of developing PD, and PIP3 represents a potent target for intervention in PD.


Assuntos
Doença de Parkinson , Humanos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Encéfalo/patologia , Lipídeos , Neurônios/patologia , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo
3.
Brain ; 143(6): 1811-1825, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32436573

RESUMO

The polyglutamine (polyQ) diseases are a group of inherited neurodegenerative diseases that include Huntington's disease, various spinocerebellar ataxias, spinal and bulbar muscular atrophy, and dentatorubral pallidoluysian atrophy. They are caused by the abnormal expansion of a CAG repeat coding for the polyQ stretch in the causative gene of each disease. The expanded polyQ stretches trigger abnormal ß-sheet conformational transition and oligomerization followed by aggregation of the polyQ proteins in the affected neurons, leading to neuronal toxicity and neurodegeneration. Disease-modifying therapies that attenuate both symptoms and molecular pathogenesis of polyQ diseases remain an unmet clinical need. Here we identified arginine, a chemical chaperone that facilitates proper protein folding, as a novel compound that targets the upstream processes of polyQ protein aggregation by stabilizing the polyQ protein conformation. We first screened representative chemical chaperones using an in vitro polyQ aggregation assay, and identified arginine as a potent polyQ aggregation inhibitor. Our in vitro and cellular assays revealed that arginine exerts its anti-aggregation property by inhibiting the toxic ß-sheet conformational transition and oligomerization of polyQ proteins before the formation of insoluble aggregates. Arginine exhibited therapeutic effects on neurological symptoms and protein aggregation pathology in Caenorhabditis elegans, Drosophila, and two different mouse models of polyQ diseases. Arginine was also effective in a polyQ mouse model when administered after symptom onset. As arginine has been safely used for urea cycle defects and for mitochondrial myopathy, encephalopathy, lactic acid and stroke syndrome patients, and efficiently crosses the blood-brain barrier, a drug-repositioning approach for arginine would enable prompt clinical application as a promising disease-modifier drug for the polyQ diseases.


Assuntos
Arginina/metabolismo , Arginina/farmacologia , Peptídeos/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Modelos Animais de Doenças , Drosophila/metabolismo , Feminino , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Doença de Huntington/genética , Masculino , Camundongos , Camundongos Endogâmicos , Chaperonas Moleculares/genética , Peptídeos/genética , Agregação Patológica de Proteínas , Conformação Proteica/efeitos dos fármacos , Dobramento de Proteína/efeitos dos fármacos , Ataxias Espinocerebelares/genética
4.
Chembiochem ; 20(16): 2151-2159, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31225928

RESUMO

Lipid packing has a strong influence on the formation and structural dynamics of cell membranes. Techniques to modulate lipid packing may thus enable modification of cellular functions and events. An 18-residue amphiphilic helical peptide derived from the N-terminal segment of epsin-1 (EpN18) is reported to induce positive membrane curvature and to loosen lipid packing in the cell membrane. In this study, it is shown that EpN18, crosslinked to a leucine-zipper peptide K4, is recruited to the cell surface by interacting with a cell-surface-expressed E3 leucine-zipper segment. Cell-surface tethering markedly enhanced loosening of lipid packing, which led to the promotion of membrane translocation of octaarginine. The loosening of lipid packing by EpN18 was also confirmed by analyzing the generalized polarization value with a membrane-environment-sensitive dye, 2-hydroxy-3-{2-[(2-hydroxyethyl)dimethylamino]ethyl}-4-{2-[6-(dibutylamino)-2-naphthyl]ethenyl}pyridiniumdibromide (di-4-ANEPPDHQ). This approach thus shows promise for the control of lipid packing and related cellular events.


Assuntos
Lipídeos/química , Peptídeos/química , Tensoativos/química , Células HeLa , Humanos , Modelos Moleculares , Estrutura Molecular , Propriedades de Superfície , Células Tumorais Cultivadas
5.
Bioconjug Chem ; 30(2): 454-460, 2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30428256

RESUMO

Marked promotion of membrane permeation of a cell-penetrating peptide, octaarginine (R8), was attained by attachment to a single 2,2'-dipicolylamine moiety (DPA-R8) that forms 1:1 complexes with metal ions. Studies using giant unilamellar vesicles demonstrated that DPA targets phospholipids and enhances R8 binding to the membranes in the presence of metal ions. While DPA/Zn(II) complex has been most frequently employed for chelate formation with phosphates, Ni(II) had the most prominent effect on the membrane binding and penetration of DPA-R8. Facile cytosolic distribution of DPA-R8 was also attained in a few minutes in the presence of Ni(II). Analysis of the cellular uptake methods of DPA-R8/Ni(II) suggested the involvement of direct permeation through cell membrane without the use of endocytosis. The applicability of this system to the intracellular delivery of bioactive compounds was exemplified using a peptidomimetic farnesyltransferase inhibitor, FTI277.


Assuntos
Peptídeos Penetradores de Células/metabolismo , Complexos de Coordenação/metabolismo , Portadores de Fármacos/metabolismo , Oligopeptídeos/metabolismo , Aminas/química , Aminas/metabolismo , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular , Peptídeos Penetradores de Células/química , Complexos de Coordenação/química , Portadores de Fármacos/química , Endocitose , Células HeLa , Humanos , Metionina/administração & dosagem , Metionina/análogos & derivados , Metionina/farmacocinética , Oligopeptídeos/química , Ácidos Picolínicos/química , Ácidos Picolínicos/metabolismo , Zinco/química , Zinco/metabolismo
6.
Biomacromolecules ; 20(5): 1849-1859, 2019 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-30893557

RESUMO

To facilitate the cytosolic delivery of larger molecules such as proteins, we developed a new cell-penetrating peptide sequence, named Pas2r12, consisting of a repeated Pas sequence (FFLIG-FFLIG) and d-dodeca-arginine (r12). This peptide significantly enhanced the cellular uptake and cytosolic release of enhanced green fluorescent protein and immunoglobulin G as cargos. We found that simply mixing Pas2r12 with cargos could generate cytosolic introducible forms. The cytosolic delivery of cargos by Pas2r12 was found to be an energy-requiring process, to rely on actin polymerization, and to be suppressed by caveolae-mediated endocytosis inhibitors (genistein and methyl-ß-cyclodextrin) and small interfering RNA against caveolin-1. These results suggest that Pas2r12 enhances membrane penetration of cargos without the need for cross-linking and that caveolae-mediated endocytosis may be the route by which cytosolic delivery is enhanced.


Assuntos
Peptídeos Penetradores de Células/metabolismo , Portadores de Fármacos/metabolismo , Endocitose , Arginina/análogos & derivados , Cavéolas/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Peptídeos Penetradores de Células/química , Citosol/metabolismo , Portadores de Fármacos/química , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Imunoglobulina G/metabolismo
7.
Biol Pharm Bull ; 41(6): 843-849, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29863073

RESUMO

Molecular chaperones have essential roles in cell survival, to prevent misfolding, aggregation, and aberrant accumulation of cellular proteins, and thus to maintain protein homeostasis (proteostasis). However, recent studies using animal models suggest that transcriptional upregulation of molecular chaperones in response to various types of stresses does not ubiquitously occur in all cells and tissues, but is a cell type-specific event. The imbalanced response to stresses between cells and tissues has been pointed out since more than 30 years ago, but the molecular basis as to how organisms maintain proteostasis in all cells, especially cells deficient for chaperone induction, remains unknown. In this review, I introduce the non-cell autonomous function of molecular chaperones that has been suggested in animal studies, especially focusing on our recent findings, and discuss the possibility that the non-cell autonomous function might provide a potential explanation as to how organisms would maintain proteostasis despite the imbalanced stress response between cells and tissues. Further elucidation of the molecular basis underlying the non-cell autonomous function of molecular chaperones would provide not only better understanding as to how organisms maintain proteostasis but also important insights into the potential development of therapies and diagnostics for the currently intractable neurodegenerative diseases that are associated with protein misfolding and aggregation.


Assuntos
Chaperonas Moleculares , Proteostase , Animais , Exossomos , Proteínas de Choque Térmico/metabolismo , Humanos
8.
Proc Natl Acad Sci U S A ; 112(19): E2497-506, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25918398

RESUMO

The heat shock response (HSR), a transcriptional response that up-regulates molecular chaperones upon heat shock, is necessary for cell survival in a stressful environment to maintain protein homeostasis (proteostasis). However, there is accumulating evidence that the HSR does not ubiquitously occur under stress conditions, but largely depends on the cell types. Despite such imbalanced HSR among different cells and tissues, molecular mechanisms by which multicellular organisms maintain their global proteostasis have remained poorly understood. Here, we report that proteostasis can be maintained by molecular chaperones not only in a cell-autonomous manner but also in a non-cell-autonomous manner. We found that elevated expression of molecular chaperones, such as Hsp40 and Hsp70, in a group of cells improves proteostasis in other groups of cells, both in cultured cells and in Drosophila expressing aggregation-prone polyglutamine proteins. We also found that Hsp40, as well as Hsp70 and Hsp90, is physiologically secreted from cells via exosomes, and that the J domain at the N terminus is responsible for its exosome-mediated secretion. Addition of Hsp40/Hsp70-containing exosomes to the culture medium of the polyglutamine-expressing cells results in efficient suppression of inclusion body formation, indicating that molecular chaperones non-cell autonomously improve the protein-folding environment via exosome-mediated transmission. Our study reveals that intercellular chaperone transmission mediated by exosomes is a novel molecular mechanism for non-cell-autonomous maintenance of organismal proteostasis that could functionally compensate for the imbalanced state of the HSR among different cells, and also provides a novel physiological role of exosomes that contributes to maintenance of organismal proteostasis.


Assuntos
Exossomos/metabolismo , Chaperonas Moleculares/metabolismo , Animais , Animais Geneticamente Modificados , Linhagem Celular , Drosophila , Drosophila melanogaster , Proteínas de Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico/metabolismo , Homeostase , Camundongos , Microscopia Eletrônica , Doenças Neurodegenerativas/patologia , Peptídeos/química , Dobramento de Proteína , Estrutura Terciária de Proteína , Transdução de Sinais , Transcrição Gênica , Transfecção
9.
Biochim Biophys Acta ; 1858(7 Pt A): 1443-50, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27039278

RESUMO

Elucidating the predominant cellular entry mechanism for protein transduction domains (PTDs) and their synthetic mimics (PTDMs) is a complicated problem that continues to be a significant source of debate in the literature. The PTDMs reported here provide a well-controlled platform to vary molecular composition for structure activity relationship studies to further our understanding of PTDs, their non-covalent association with cargo, and their cellular internalization pathways. Specifically, several guanidine rich homopolymers, along with an amphiphilic block copolymer were used to investigate the relationship between structure and internalization activity in HeLa cells, both alone and non-covalently complexed with EGFP by flow cytometery and confocal imaging. The findings indicate that while changing the amount of positive charge on our PTDMs does not seem to affect the endosomal uptake, the presence of hydrophobicity appears to be a critical factor for the polymers to enter cells either alone, or with associated cargo.


Assuntos
Endocitose/fisiologia , Endossomos/metabolismo , Guanidinas/metabolismo , Peptidomiméticos/metabolismo , Polímeros/metabolismo , Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/metabolismo , Citometria de Fluxo , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Guanidinas/síntese química , Células HeLa , Humanos , Interações Hidrofóbicas e Hidrofílicas , Peptidomiméticos/síntese química , Polímeros/síntese química , Estrutura Terciária de Proteína , Transporte Proteico , Eletricidade Estática , Relação Estrutura-Atividade
10.
Hum Mol Genet ; 24(23): 6675-86, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26362253

RESUMO

Alpha-synuclein (αSyn) plays a central role in the pathogenesis of Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Recent multicenter genetic studies have revealed that mutations in the glucocerebrosidase 1 (GBA1) gene, which are responsible for Gaucher's disease, are strong risk factors for PD and DLB. However, the mechanistic link between the functional loss of glucocerebrosidase (GCase) and the toxicity of αSyn in vivo is not fully understood. In this study, we employed Drosophila models to examine the effect of GCase deficiency on the neurotoxicity of αSyn and its molecular mechanism. Behavioral and histological analyses showed that knockdown of the Drosophila homolog of GBA1 (dGBA1) exacerbates the locomotor dysfunction, loss of dopaminergic neurons and retinal degeneration of αSyn-expressing flies. This phenotypic aggravation was associated with the accumulation of proteinase K (PK)-resistant αSyn, rather than with changes in the total amount of αSyn, raising the possibility that glucosylceramide (GlcCer), a substrate of GCase, accelerates the misfolding of αSyn. Indeed, in vitro experiments revealed that GlcCer directly promotes the conversion of recombinant αSyn into the PK-resistant form, representing a toxic conformational change. Similar to dGBA1 knockdown, knockdown of the Drosophila homolog of ß-galactosidase (ß-Gal) also aggravated locomotor dysfunction of the αSyn flies, and its substrate GM1 ganglioside accelerated the formation of PK-resistant αSyn. Our findings suggest that the functional loss of GCase or ß-Gal promotes the toxic conversion of αSyn via aberrant interactions between αSyn and their substrate glycolipids, leading to the aggravation of αSyn-mediated neurodegeneration.


Assuntos
Glucosilceramidase/genética , Transtornos Parkinsonianos/etiologia , alfa-Sinucleína/metabolismo , Animais , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Endopeptidase K/metabolismo , Técnicas de Silenciamento de Genes , Glucosilceramidase/deficiência , Glucosilceramidase/metabolismo , Glucosilceramidas/metabolismo , Humanos , Masculino , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/fisiopatologia , Agregação Patológica de Proteínas , Dobramento de Proteína , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
11.
Biopolymers ; 108(1)2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27554421

RESUMO

Calmodulin is a representative calcium-binding protein comprised of four Ca2+ -binding motifs with a helix-loop-helix structure (EF-hands). In this study, we clarified the potential of peptide segments derived from the third and fourth EF-hands (EF3 and EF4) to act as recognition tags. Through an analysis of the mode of disulfide formation among cysteines inserted at the N- or C-terminus of these peptide segments, EF3 and EF4 peptides were suggested to form a heterodimer with a topology similar to that in the wild-type protein. Heterodimer formation was shown to be a function of the Ca2+ concentration, suggesting that these structures may be used as Ca2+ -switchable recognition tags. An example of an "EF-tag" system involving the membrane fusion of liposomes decorated with EF3 and EF4 peptides is presented. © 2016 Wiley Periodicals, Inc. Biopolymers (Pept Sci), 2016.


Assuntos
Cálcio/química , Calmodulina/química , Peptídeos/química , Sequência de Aminoácidos , Dimerização , Motivos EF Hand , Lipossomos/química , Lipossomos/metabolismo , Dados de Sequência Molecular , Oxirredução , Peptídeos/síntese química , Peptídeos/metabolismo , Estrutura Terciária de Proteína
12.
Bioconjug Chem ; 27(4): 1119-30, 2016 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-27019270

RESUMO

Arginine-rich cell-penetrating peptides (CPPs) such as Tat and oligoarginine peptides have been widely used as carriers for intracellular delivery of bioactive molecules. Despite accumulating evidence for involvement of endocytosis in the cellular uptake of arginine-rich CPPs, the primary cell-surface receptors for these peptide carriers that would initiate endocytic processes leading to intracellular delivery of bioactive cargoes have remained poorly understood. Our previous attempt to identify membrane receptors for octa-arginine (R8) peptide, one of the representative arginine-rich CPPs, using the photo-cross-linking probe bearing a photoreactive diazirine was not successful due to considerable amounts of cellular proteins nonspecifically bound to the affinity beads. To address this issue, here we developed a photo-cross-linking probe in which a cleavable linker of a diazobenzene moiety was employed to allow selective elution of cross-linked proteins by reducing agent-mediated cleavage. We demonstrated that introduction of the diazobenzene moiety into the photoaffinity probe enables efficient purification of cross-linked proteins with significant reduction of nonspecific binding proteins, leading to successful identification of 17 membrane-associated proteins that would interact with R8 peptide. RNAi-mediated knockdown experiments in combination with the pharmacological inhibitors revealed that, among the proteins identified, syndecan-4, one of the heparan sulfate proteoglycans, is an endogenous membrane-associated receptor for the cellular uptake of R8 peptide via clathrin-mediated endocytosis. This syndecan-4-dependent pathway was also involved in the intracellular delivery of bioactive proteins mediated by R8 peptide. These results reveal that syndecan-4 is a primary cell-surface target for R8 peptide that allows intracellular delivery of bioactive cargo molecules via clathrin-mediated endocytosis.


Assuntos
Arginina/metabolismo , Endocitose/fisiologia , Sindecana-4/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Sindecana-4/fisiologia
13.
Chem Pharm Bull (Tokyo) ; 64(10): 1431-1437, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27725497

RESUMO

Arginine-rich cell-penetrating peptides (CPPs) including Tat, Penetratin and oligoarginine peptides are a series of short peptides that can be efficiently internalized into cells and have been widely used as carriers for intracellular delivery of bioactive molecules. In the early phase of the study, CPPs, as well as their conjugates, were thought to rapidly enter cells by direct penetration through membranes, which was later found to be an experimental artifact that was concluded from observations in fixed cells. Although re-evaluation using living unfixed cells revealed that endocytosis has a major role in internalization of these peptides, there are a number of studies reporting that, even if fixation is avoided, direct translocation across plasma membranes and cytosolic distribution of arginine-rich CPPs are still observed in cells without membrane perturbation. In addition, amphiphilic counteranions such as pyrenebutyrate dramatically accelerate direct translocation of these peptides into cells. These results suggest that there are at least two pathways, i.e., endocytosis and direct translocation, both of which would contribute to cellular internalization of arginine-rich CPPs. In this review, we first introduce the story of fixation artifact, which indeed led to the critical progress in CPP study, and then summarize the current understanding for direct translocation of arginine-rich CPPs. Comprehensive understanding of direct translocation of these peptides and its mechanistic elucidation would provide useful knowledge for developing methodologies that would enable efficient intracellular delivery.


Assuntos
Arginina/metabolismo , Peptídeos Penetradores de Células/metabolismo , Animais
14.
Nature ; 458(7234): 106-9, 2009 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-19262675

RESUMO

In-cell NMR is an isotope-aided multi-dimensional NMR technique that enables observations of conformations and functions of proteins in living cells at the atomic level. This method has been successfully applied to proteins overexpressed in bacteria, providing information on protein-ligand interactions and conformations. However, the application of in-cell NMR to eukaryotic cells has been limited to Xenopus laevis oocytes. Wider application of the technique is hampered by inefficient delivery of isotope-labelled proteins into eukaryote somatic cells. Here we describe a method to obtain high-resolution two-dimensional (2D) heteronuclear NMR spectra of proteins inside living human cells. Proteins were delivered to the cytosol by the pyrenebutyrate-mediated action of cell-penetrating peptides linked covalently to the proteins. The proteins were subsequently released from cell-penetrating peptides by endogenous enzymatic activity or by autonomous reductive cleavage. The heteronuclear 2D spectra of three different proteins inside human cells demonstrate the broad application of this technique to studying interactions and protein processing. The in-cell NMR spectra of FKBP12 (also known as FKBP1A) show the formation of specific complexes between the protein and extracellularly administered immunosuppressants, demonstrating the utility of this technique in drug screening programs. Moreover, in-cell NMR spectroscopy demonstrates that ubiquitin has much higher hydrogen exchange rates in the intracellular environment, possibly due to multiple interactions with endogenous proteins.


Assuntos
Espaço Intracelular/metabolismo , Ressonância Magnética Nuclear Biomolecular/métodos , Proteínas Recombinantes de Fusão/química , Animais , Permeabilidade da Membrana Celular , Sobrevivência Celular/efeitos dos fármacos , Medição da Troca de Deutério , Avaliação Pré-Clínica de Medicamentos/métodos , Produtos do Gene tat/genética , Produtos do Gene tat/metabolismo , Células HeLa , Humanos , Imunossupressores/química , Imunossupressores/metabolismo , Imunossupressores/farmacologia , Ligação Proteica , Pirenos/farmacologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína 1A de Ligação a Tacrolimo/química , Proteína 1A de Ligação a Tacrolimo/genética , Proteína 1A de Ligação a Tacrolimo/metabolismo , Transfecção , Ubiquitina/genética , Ubiquitina/metabolismo
15.
Acta Neuropathol Commun ; 12(1): 20, 2024 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-38311779

RESUMO

The abnormal aggregation of TDP-43 into cytoplasmic inclusions in affected neurons is a major pathological hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Although TDP-43 is aberrantly accumulated in the neurons of most patients with sporadic ALS/FTD and other TDP-43 proteinopathies, how TDP-43 forms cytoplasmic aggregates remains unknown. In this study, we show that a deficiency in DCTN1, a subunit of the microtubule-associated motor protein complex dynactin, perturbs the dynamics of stress granules and drives the formation of TDP-43 cytoplasmic aggregation in cultured cells, leading to the exacerbation of TDP-43 pathology and neurodegeneration in vivo. We demonstrated using a Drosophila model of ALS/FTD that genetic knockdown of DCTN1 accelerates the formation of ubiquitin-positive cytoplasmic inclusions of TDP-43. Knockdown of components of other microtubule-associated motor protein complexes, including dynein and kinesin, also increased the formation of TDP-43 inclusions, indicating that intracellular transport along microtubules plays a key role in TDP-43 pathology. Notably, DCTN1 knockdown delayed the disassembly of stress granules in stressed cells, leading to an increase in the formation of pathological cytoplasmic inclusions of TDP-43. Our results indicate that a deficiency in DCTN1, as well as disruption of intracellular transport along microtubules, is a modifier that drives the formation of TDP-43 pathology through the dysregulation of stress granule dynamics.


Assuntos
Esclerose Lateral Amiotrófica , Proteínas de Ligação a DNA , Proteínas de Drosophila , Complexo Dinactina , Demência Frontotemporal , Animais , Humanos , Esclerose Lateral Amiotrófica/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Drosophila/metabolismo , Complexo Dinactina/genética , Demência Frontotemporal/patologia , Grânulos de Estresse , Proteínas de Drosophila/genética
16.
Mol Ther ; 20(5): 984-93, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22334015

RESUMO

Endocytosis has been implicated in the cellular uptake of arginine-rich, cell-penetrating peptides (CPPs). However, accumulating evidence suggests that certain conditions allow the direct, non-endocytic penetration of arginine-rich peptides through the plasma membrane. We previously showed that Alexa Fluor 488-labeled dodeca-arginine (R12-Alexa488) directly enters cells at specific sites on the plasma membrane and subsequently diffuses throughout cells. In this study, we found that the peptide influx was accompanied by the formation of unique, "particle-like" multivesicular structures on the plasma membrane, together with topical inversion of the plasma membrane. Importantly, the conjugation of dodeca-arginine (R12) to Alexa Fluor 488 or a peptide tag derived from hemagglutinin (HAtag) significantly accelerated particle formation, suggesting that the chemical properties of the attached molecules (cargo molecules) may contribute to translocation of the R12 peptide. Coincubation with R12-HAtag allowed the membrane-impermeable R4-Alexa488 to permeate cells. These results suggest that R12 peptides attached to hydrophobic cargo molecules stimulate dynamic morphological alterations in the plasma membrane, and that these structural changes allow the peptides to permeate the plasma membrane. These findings may provide a novel mode of cell permeabilization by arginine-rich peptides as a means of drug delivery.


Assuntos
Arginina/química , Membrana Celular/ultraestrutura , Peptídeos Penetradores de Células/síntese química , Oligopeptídeos/síntese química , Arginina/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/metabolismo , Peptídeos Penetradores de Células/farmacologia , Difusão , Corantes Fluorescentes , Células HeLa , Hemaglutininas/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Maleimidas , Microscopia Confocal , Microscopia Eletrônica , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Transporte Proteico
17.
Sci Rep ; 13(1): 17801, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37853169

RESUMO

Cerebellar ataxia, neuropathy, and vestibular areflexia syndrome (CANVAS) has recently been attributed to biallelic repeat expansions in RFC1. More recently, the disease entity has expanded to atypical phenotypes, including chronic neuropathy without cerebellar ataxia or vestibular areflexia. Very recently, RFC1 expansions were found in patients with Sjögren syndrome who had neuropathy that did not respond to immunotherapy. In this study RFC1 was examined in 240 patients with acute or chronic neuropathies, including 105 with Guillain-Barré syndrome or Miller Fisher syndrome, 76 with chronic inflammatory demyelinating polyneuropathy, and 59 with other types of chronic neuropathy. Biallelic RFC1 mutations were found in three patients with immune-mediated neuropathies, including Guillain-Barré syndrome, idiopathic sensory ataxic neuropathy, or anti-myelin-associated glycoprotein (MAG) neuropathy, who responded to immunotherapies. In addition, a patient with chronic sensory autonomic neuropathy had biallelic mutations, and subclinical changes in Schwann cells on nerve biopsy. In summary, we found CANVAS-related RFC1 mutations in patients with treatable immune-mediated neuropathy or demyelinating neuropathy.


Assuntos
Vestibulopatia Bilateral , Ataxia Cerebelar , Síndrome de Guillain-Barré , Doenças do Sistema Nervoso Periférico , Doenças Vestibulares , Humanos , Ataxia Cerebelar/genética , Mutação , Doenças do Sistema Nervoso Periférico/genética
18.
Mol Ther Nucleic Acids ; 31: 353-366, 2023 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-36817728

RESUMO

The abnormal aggregation of TDP-43 into cytoplasmic inclusions in affected neurons is a pathological hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Although how TDP-43 forms cytoplasmic aggregates and causes neurodegeneration in patients with ALS/FTD remains unclear, reducing cellular TDP-43 levels is likely to prevent aggregation and to rescue neurons from TDP-43 toxicity. To address this issue, here we developed gapmer-type antisense oligonucleotides (ASOs) against human TDP-43 using 2'-O,4'-C-ethylene nucleic acids (ENAs), which are modified nucleic acids with high stability, and tested the therapeutic potential of lowering TDP-43 levels using ENA-modified ASOs. We demonstrated that intracerebroventricular administration of ENA-modified ASOs into a mouse model of ALS/FTD expressing human TDP-43 results in the efficient reduction of TDP-43 levels in the brain and spinal cord. Surprisingly, a single injection of ENA-modified ASOs into TDP-43 mice led to long-lasting improvement of behavioral abnormalities and the suppression of cytoplasmic TDP-43 aggregation, even after TDP-43 levels had returned to the initial levels. Our results demonstrate that transient reduction of TDP-43 using ENA-modified ASOs leads to sustained therapeutic benefits in vivo, indicating the possibility of a disease-modifying therapy by lowering TDP-43 levels for the treatment of the TDP-43 proteinopathies, including ALS/FTD.

19.
Elife ; 122023 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-37461319

RESUMO

Abnormal expansions of GGGGCC repeat sequence in the noncoding region of the C9orf72 gene is the most common cause of familial amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD). The expanded repeat sequence is translated into dipeptide repeat proteins (DPRs) by noncanonical repeat-associated non-AUG (RAN) translation. Since DPRs play central roles in the pathogenesis of C9-ALS/FTD, we here investigate the regulatory mechanisms of RAN translation, focusing on the effects of RNA-binding proteins (RBPs) targeting GGGGCC repeat RNAs. Using C9-ALS/FTD model flies, we demonstrated that the ALS/FTD-linked RBP FUS suppresses RAN translation and neurodegeneration in an RNA-binding activity-dependent manner. Moreover, we found that FUS directly binds to and modulates the G-quadruplex structure of GGGGCC repeat RNA as an RNA chaperone, resulting in the suppression of RAN translation in vitro. These results reveal a previously unrecognized regulatory mechanism of RAN translation by G-quadruplex-targeting RBPs, providing therapeutic insights for C9-ALS/FTD and other repeat expansion diseases.


Assuntos
Esclerose Lateral Amiotrófica , Demência Frontotemporal , Humanos , Esclerose Lateral Amiotrófica/patologia , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Demência Frontotemporal/patologia , RNA/metabolismo , Proteína FUS de Ligação a RNA/genética , Proteínas de Ligação a RNA/genética , Drosophila/genética
20.
Neurochem Int ; 157: 105357, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35525394

RESUMO

Polyglutamine (PolyQ) diseases are a group of inherited neurodegenerative diseases including Huntington's disease and several types of spinocerebellar ataxias, which are caused by aggregation and accumulation of the disease-causative proteins with an abnormally expanded PolyQ stretch. Extracellular vesicles (EVs) are membrane particles that are released from cells, including exosomes, microvesicles, and other extracellular particles. Recent studies have suggested that the PolyQ proteins, which are the disease-causative proteins of PolyQ diseases, and its aggregates are secreted via EVs, similar to the aggregation-prone proteins associated with other neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. The PolyQ proteins that are secreted from cells can transmit intercellularly, which may contribute to pathological propagation of the PolyQ protein aggregates in patient brain, and therefore, the pathological roles of EVs in the onset and progression of PolyQ diseases has attracted much attention. EVs may also mediate intercellular transfer of heat shock proteins and other neuroprotective factors, which are beneficial for protein homeostasis and cell survival, and thus, have therapeutic potential for the neurodegenerative diseases including PolyQ diseases. Furthermore, because EVs contain not only the disease-associated proteins, but also various proteins, miRNAs and other components, and changes in the levels of these contents might reflect pathological changes, EVs derived from blood, cerebrospinal fluid, and urine would be a potential source of minimally invasive diagnostic biomarkers that report disease-associated changes in PolyQ diseases. In this review, we summarize the current understanding of the pathological roles of EVs in PolyQ diseases, and therapeutic and diagnostic potential of EVs for these diseases. Elucidation of the pathological and physiological roles of EVs would lead to identification of a proper therapeutic target that would not interfere the protective roles of EVs for cell survival but suppress pathological propagation of the disease-causative proteins in PolyQ disease.


Assuntos
Vesículas Extracelulares , Ataxias Espinocerebelares , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Humanos , Proteínas Mutantes/metabolismo , Peptídeos/metabolismo , Ataxias Espinocerebelares/diagnóstico , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA