Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Nat Commun ; 14(1): 2683, 2023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-37160917

RESUMO

Many secretory enzymes acquire essential zinc ions (Zn2+) in the Golgi complex. ERp44, a chaperone operating in the early secretory pathway, also binds Zn2+ to regulate its client binding and release for the control of protein traffic and homeostasis. Notably, three membrane transporter complexes, ZnT4, ZnT5/ZnT6 and ZnT7, import Zn2+ into the Golgi lumen in exchange with protons. To identify their specific roles, we here perform quantitative Zn2+ imaging using super-resolution microscopy and Zn2+-probes targeted in specific Golgi subregions. Systematic ZnT-knockdowns reveal that ZnT4, ZnT5/ZnT6 and ZnT7 regulate labile Zn2+ concentration at the distal, medial, and proximal Golgi, respectively, consistent with their localization. Time-course imaging of cells undergoing synchronized secretory protein traffic and functional assays demonstrates that ZnT-mediated Zn2+ fluxes tune the localization, trafficking, and client-retrieval activity of ERp44. Altogether, this study provides deep mechanistic insights into how ZnTs control Zn2+ homeostasis and ERp44-mediated proteostasis along the early secretory pathway.


Assuntos
Complexo de Golgi , Proteostase , Humanos , Homeostase , Transporte Biológico , Bioensaio , Proteínas de Membrana , Chaperonas Moleculares
3.
ACS Sens ; 7(3): 748-757, 2022 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-35238552

RESUMO

Although many Zn2+ fluorescent probes have been developed, there remains a lack of consensus on the labile Zn2+ concentrations ([Zn2+]) in several cellular compartments, as the fluorescence properties and zinc affinity of the fluorescent probes are greatly affected by the pH and redox environments specific to organelles. In this study, we developed two turn-on-type Zn2+ fluorescent probes, namely, ZnDA-2H and ZnDA-3H, with low pH sensitivity and suitable affinity (Kd = 5.0 and 0.16 nM) for detecting physiological labile Zn2+ in various cellular compartments, such as the cytosol, nucleus, ER, and mitochondria. Due to their sufficient membrane permeability, both probes were precisely localized to the target organelles in HeLa cells using HaloTag labeling technology. Using an in situ standard quantification method, we identified the [Zn2+] in the tested organelles, resulting in the subcellular [Zn2+] distribution as [Zn2+]ER < [Zn2+]mito < [Zn2+]cyto ∼ [Zn2+]nuc.


Assuntos
Corantes Fluorescentes , Zinco , Núcleo Celular , Corantes Fluorescentes/química , Células HeLa , Humanos , Microscopia de Fluorescência , Zinco/química
4.
Structure ; 29(12): 1357-1370.e6, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33857433

RESUMO

P5, also known as PDIA6, is a PDI family member involved in the ER quality control. Here, we revealed that P5 dimerizes via a unique adhesive motif contained in the N-terminal thioredoxin-like domain. Unlike conventional leucine zipper motifs with leucine residues every two helical turns on ∼30-residue parallel α helices, this adhesive motif includes periodic repeats of leucine/valine residues at the third or fourth position spanning five helical turns on 15-residue anti-parallel α helices. The P5 dimerization interface is further stabilized by several reciprocal salt bridges and C-capping interactions between protomers. A monomeric P5 mutant with the impaired adhesive motif showed structural instability and local unfolding, and behaved as aberrant proteins that induce the ER stress response. Disassembly of P5 to monomers compromised its ability to inactivate IRE1α via intermolecular disulfide bond reduction and its Ca2+-dependent regulation of chaperone function in vitro. Thus, the leucine-valine adhesive motif supports structure and function of P5.


Assuntos
Leucina/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Valina/metabolismo , Dimerização , Humanos , Estrutura Molecular , Dobramento de Proteína
5.
Cell Chem Biol ; 27(12): 1521-1531.e8, 2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-32997976

RESUMO

Fluorescent Zn2+ probes used for the quantitative analysis of labile Zn2+ concentration ([Zn2+]) in target organelles are crucial for understanding the role of Zn2+ in biological processes. Although several fluorescent Zn2+ probes have been developed to date, there is still a lack of consensus concerning the [Zn2+] in intracellular organelles. In this study, we describe the development of ZnDA-1H, a small-molecule fluorescent probe for Zn2+, which exhibits less pH sensitivity, high Zn2+ selectivity, and large fluorescence enhancement upon binding to Zn2+. Through protein labeling technology, ZnDA-1H was precisely targeted in various intracellular organelles, such as the nucleus, mitochondria, endoplasmic reticulum, and Golgi apparatus. ZnDA-1H exhibited a reversible fluorescence response toward labile Zn2+ in these organelles in live cells. Using this probe, the [Zn2+] in the Golgi apparatus was estimated to be 25 ± 1 nM, suggesting that labile Zn2+ plays a physiological role in the secretory pathway.


Assuntos
Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Complexo de Golgi/metabolismo , Microscopia de Fluorescência , Zinco/metabolismo , Células HeLa , Humanos , Coloração e Rotulagem
6.
Nat Commun ; 10(1): 603, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30723194

RESUMO

Zinc ions (Zn2+) are imported into the early secretory pathway by Golgi-resident transporters, but their handling and functions are not fully understood. Here, we show that Zn2+ binds with high affinity to the pH-sensitive chaperone ERp44, modulating its localization and ability to retrieve clients like Ero1α and ERAP1 to the endoplasmic reticulum (ER). Silencing the Zn2+ transporters that uptake Zn2+ into the Golgi led to ERp44 dysfunction and increased secretion of Ero1α and ERAP1. High-resolution crystal structures of Zn2+-bound ERp44 reveal that Zn2+ binds to a conserved histidine-cluster. The consequent large displacements of the regulatory C-terminal tail expose the substrate-binding surface and RDEL motif, ensuring client capture and retrieval. ERp44 also forms Zn2+-bridged homodimers, which dissociate upon client binding. Histidine mutations in the Zn2+-binding sites compromise ERp44 activity and localization. Our findings reveal a role of Zn2+ as a key regulator of protein quality control at the ER-Golgi interface.


Assuntos
Proteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Via Secretória , Zinco/metabolismo , Aminopeptidases/metabolismo , Sítios de Ligação/genética , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Cristalografia por Raios X , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Células HeLa , Células Hep G2 , Humanos , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Antígenos de Histocompatibilidade Menor/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Oxirredutases/metabolismo , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Controle de Qualidade , Interferência de RNA , Zinco/química
7.
Structure ; 25(6): 846-857.e4, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28479060

RESUMO

ERdj5, composed of an N-terminal J domain followed by six thioredoxin-like domains, is the largest protein disulfide isomerase family member and functions as an ER-localized disulfide reductase that enhances ER-associated degradation (ERAD). Our previous studies indicated that ERdj5 comprises two regions, the N- and C-terminal clusters, separated by a linker loop and with distinct functional roles in ERAD. We here present a new crystal structure of ERdj5 with a largely different cluster arrangement relative to that in the original crystal structure. Single-molecule observation by high-speed atomic force microscopy visualized rapid cluster movement around the flexible linker loop, indicating the highly dynamic nature of ERdj5 in solution. ERdj5 mutants with a fixed-cluster orientation compromised the ERAD enhancement activity, likely because of less-efficient reduction of aberrantly formed disulfide bonds and prevented substrate transfer in the ERdj5-mediated ERAD pathway. We propose a significant role of ERdj5 conformational dynamics in ERAD of disulfide-linked oligomers.


Assuntos
Degradação Associada com o Retículo Endoplasmático/fisiologia , Proteínas de Choque Térmico HSP40/química , Proteínas de Choque Térmico HSP40/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Cristalografia por Raios X , Dissulfetos/química , Dissulfetos/metabolismo , Chaperona BiP do Retículo Endoplasmático , Proteínas de Choque Térmico HSP40/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Microscopia de Força Atômica , Modelos Moleculares , Chaperonas Moleculares/genética , Mutação , Conformação Proteica
8.
Angew Chem Int Ed Engl ; 56(20): 5522-5526, 2017 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-28394477

RESUMO

Synthetic insulin analogues with a long lifetime are current drug targets for the therapy of diabetic patients. The replacement of the interchain disulfide with a diselenide bridge, which is more resistant to reduction and internal bond rotation, can enhance the lifetime of insulin in the presence of the insulin-degrading enzyme (IDE) without impairing the hormonal function. The [C7UA ,C7UB ] variant of bovine pancreatic insulin (BPIns) was successfully prepared by using two selenocysteine peptides (i.e., the C7U analogues of A- and B-chains, respectively). In a buffer solution at pH 10 they spontaneously assembled under thermodynamic control to the correct insulin fold. The selenoinsulin (Se-Ins) exhibited a bioactivity comparable to that of BPIns. Interestingly, degradation of Se-Ins with IDE was significantly decelerated (τ1/2 ≈8 h vs. ≈1 h for BPIns). The lifetime enhancement could be due to both the intrinsic stability of the diselenide bond and local conformational changes induced by the substitution.


Assuntos
Insulina/química , Insulina/síntese química , Sequência de Aminoácidos , Cristalografia por Raios X , Dissulfetos/química , Insulina/análogos & derivados , Modelos Moleculares
9.
J Biochem ; 158(2): 139-53, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25787272

RESUMO

Macroautophagy is a bulk degradation system conserved from yeast to human. In budding yeast, the guanine nucleotide-exchange factor (GEF) Sec2p is required for autophagy. We examined the role of Rabin8 (a mammalian ortholog of Sec2p) with Rab8-GEF activity in autophagy in mammalian cells. Unexpectedly, depletion of Rabin8 promoted nutrient starvation-induced autophagosome formation, indicating that Rabin8 suppresses autophagosome formation. Depletion of Rab8 did not affect autophagosome formation, and expression of a Rabin8 GEF-domain mutant reverted the Rabin8 depletion-induced increase in autophagosomes, indicating that Rabin8 suppresses autophagosome formation independently of its Rab8-GEF activity. Nuclear Dbf2-related (NDR) kinases phosphorylate Rabin8 at Ser-272. The non-phosphorylatable Rabin8-S272A mutant did not revert the Rabin8 depletion-induced increase in autophagosomes, suggesting that Ser-272 phosphorylation of Rabin8 is involved in its suppressive function in autophagy. Depletion of NDR kinases enhanced autophagosome formation and reduced mammalian/mechanistic target of rapamycin complex 1 (mTORC1) activity, suggesting that NDR kinases suppress autophagosome formation by increasing mTORC1 activity, in addition to phosphorylating Rabin8. Expression of a C-terminal fragment of Rabin8, but not that of Sec2p, suppressed nutrient starvation-induced autophagosome formation. Thus, contrary to the stimulative role of yeast Sec2p, Rabin8 has a suppressive function in autophagy in mammalian cells through its non-conserved C-terminal region.


Assuntos
Autofagia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fagossomos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Quinases do Centro Germinativo , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/química , RNA Interferente Pequeno/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Serina-Treonina Quinases TOR/metabolismo
10.
EMBO J ; 32(6): 874-85, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-23435566

RESUMO

Primary cilia are antenna-like sensory organelles protruding from the plasma membrane. Defects in ciliogenesis cause diverse genetic disorders. NDR2 was identified as the causal gene for a canine ciliopathy, early retinal degeneration, but its role in ciliogenesis remains unknown. Ciliary membranes are generated by transport and fusion of Golgi-derived vesicles to the pericentrosome, a process requiring Rab11-mediated recruitment of Rabin8, a GDP-GTP exchange factor (GEF) for Rab8, and subsequent Rab8 activation and Rabin8 binding to Sec15, a component of the exocyst that mediates vesicle tethering. This study shows that NDR2 phosphorylates Rabin8 at Ser-272 and defects in this phosphorylation impair preciliary membrane assembly and ciliogenesis, resulting in accumulation of Rabin8-/Rab11-containing vesicles at the pericentrosome. Rabin8 binds to and colocalizes with GTP-bound Rab11 and phosphatidylserine (PS) on pericentrosomal vesicles. The phospho-mimetic S272E mutation of Rabin8 decreases affinity for PS but increases affinity for Sec15. These results suggest that NDR2-mediated Rabin8 phosphorylation is crucial for ciliogenesis by triggering the switch in binding specificity of Rabin8 from PS to Sec15, thereby promoting local activation of Rab8 and ciliary membrane formation.


Assuntos
Cílios/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Fosfatidilserinas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Diferenciação Celular , Membrana Celular/metabolismo , Cílios/genética , Cílios/metabolismo , Cães , Quinases do Centro Germinativo , Humanos , Fosforilação/fisiologia , Ligação Proteica , Células Sf9 , Spodoptera , Especificidade por Substrato/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA